US11447539B2 - PD-L1 binding fibronectin type III domains - Google Patents

PD-L1 binding fibronectin type III domains Download PDF

Info

Publication number
US11447539B2
US11447539B2 US16/801,787 US202016801787A US11447539B2 US 11447539 B2 US11447539 B2 US 11447539B2 US 202016801787 A US202016801787 A US 202016801787A US 11447539 B2 US11447539 B2 US 11447539B2
Authority
US
United States
Prior art keywords
seq
protein
domain
specifically binds
amino acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US16/801,787
Other versions
US20210024612A1 (en
Inventor
Michael Diem
Rebecca Hawkins
Steven Jacobs
Manuel Sepulveda
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Janssen Biotech Inc
Original Assignee
Janssen Biotech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Janssen Biotech Inc filed Critical Janssen Biotech Inc
Priority to US16/801,787 priority Critical patent/US11447539B2/en
Assigned to JANSSEN BIOTECH, INC. reassignment JANSSEN BIOTECH, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DIEM, MICHAEL, JACOBS, STEVEN, HAWKINS, Rebecca, SEPULVEDA, MANUEL
Publication of US20210024612A1 publication Critical patent/US20210024612A1/en
Application granted granted Critical
Publication of US11447539B2 publication Critical patent/US11447539B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/78Connective tissue peptides, e.g. collagen, elastin, laminin, fibronectin, vitronectin, cold insoluble globulin [CIG]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70596Molecules with a "CD"-designation not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/179Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2318/00Antibody mimetics or scaffolds
    • C07K2318/20Antigen-binding scaffold molecules wherein the scaffold is not an immunoglobulin variable region or antibody mimetics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/40Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation
    • C07K2319/41Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation containing a Myc-tag
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/55Fusion polypeptide containing a fusion with a toxin, e.g. diphteria toxin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/60Fusion polypeptide containing spectroscopic/fluorescent detection, e.g. green fluorescent protein [GFP]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/61Fusion polypeptide containing an enzyme fusion for detection (lacZ, luciferase)

Definitions

  • the present invention relates to fibronectin type III domains that specifically bind to PD-L1 and methods of making and using the molecules.
  • the immune system is tightly controlled by a network of costimulatory and co-inhibitory ligands and receptors. These molecules provide secondary signals for T cell activation and provide a balanced network of positive and negative signals to maximize immune responses against infection and tumors, while limiting immunity to self (Wang et al., (Epub Mar. 7, 2011) J Exp Med 208(3):577-92; Lepenies et al., (2008) Endocr Metab Immune Disord Drug Targets 8:279-288).
  • Programmed Death-1 is a key immune checkpoint receptor expressed by activated T and B cells and mediates immunosuppression.
  • the ligand for PD-1, PD-L1 is expressed by antigen-presenting cells and many cancers such as lung, ovarian and colon carcinoma and various myelomas. Binding of PD-L1 to PD-1 on T cells downregulates T cell proliferation and activation and drives T cell anergy and exhaustion in the tumor microenvironment, facilitating tumor cell escape from T-cell mediated immune surveillance.
  • Measuring the expression of PD-L1 protein in the tumor tissue may aid in the early detection of cancer pathologies and may help assess the efficacy and durability of PD-L1 and PD-1 antagonists.
  • PD-L1 expression in at least 50% of tumor cells correlated with improved efficacy of Keytruda® (pembrolizumab) (Garon et al., N Engl J Med 2015; 372:2018-2028), and PD-L1 expression has been correlated with poor prognosis (see for example Wang et al., Eur J Surg oncol 2015 Apr; 41(4):450-6).
  • PD-L1 protein expression as an accurate predictor for cancer and/or the efficacy of anti-PD-1 and anti-PD-L1 directed therapies remain challenging partially due to observed variability in results depending on the detection reagent used.
  • the evaluation of PD-L1 expression in non-small cell lung cancer samples using commercially available assays such as PD-L1 (E1L3NO) XP® Rabbit mAb (Cell Signaling) and Ventana PD-L1 (SP142) Assay yielded discordant results (McLaughlin et al., JAMA Oncol 2016 Jan;2(1):46-54)
  • the invention provides an isolated FN3 domain that specifically binds to PD-L1.
  • the invention also provides an isolated FN3 domain that specifically binds to PD-L1 comprising the sequence of SEQ ID NOs: 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120,
  • the invention also provides an isolated polynucleotide encoding the FN3 domain that specifically binds to PD-L1 of the invention.
  • the invention also provides a vector comprising the polynucleotide of the invention.
  • the invention also provides a host cell comprising the vector of the invention.
  • the invention also provides a method of producing the FN3 domain that specifically binds to PD-L1 of the invention, comprising culturing the isolated host cell of the invention under conditions that the FN3 domain that specifically binds to PD-L1 is expressed, and purifying the FN3 domain that specifically binds to PD-L1.
  • the invention also provides a pharmaceutical composition comprising the FN3 domain that specifically binds to PD-L1 of the invention and a pharmaceutically acceptable carrier.
  • the invention also provides an anti-idiotypic antibody that specifically binds the FN3 domain that specifically binds to PD-L1 of the invention.
  • the invention also provides a kit comprising the FN3 domain of the invention.
  • the invention also provides a method of detecting PD-L1-expressing cancer cells in a tumor tissue, comprising
  • the invention also provides a method of isolating or detecting PD-L1 expressing cells, comprising obtaining a sample from a subject;
  • the invention also provides a method of detecting PD-L1-expressing cancer cells in a tumor tissue, comprising
  • Fibronectin type III (FN3) domain refers to a domain occurring frequently in proteins including fibronectins, tenascin, intracellular cytoskeletal proteins, cytokine receptors and prokaryotic enzymes (Bork and Doolittle, Proc Nat Acad Sci USA 89:8990-8994, 1992; Meinke et al., J Bacteriol 175:1910-1918, 1993; Watanabe et al., J Biol Chem 265:15659-15665, 1990).
  • Exemplary FN3 domains are the 15 different FN3 domains present in human tenascin C, the 15 different FN3 domains present in human fibronectin (FN), and non-natural synthetic FN3 domains as described for example in U.S. Pat. No. 8,278,419.
  • Individual FN3 domains are referred to by domain number and protein name, e.g., the 3 rd FN3 domain of tenascin (TN3), or the 10 th FN3 domain of fibronectin (FN10).
  • Cyrin refers to a FN3 domain that is based on the consensus sequence of the 15 different FN3 domains present in human tenascin C.
  • capture agent refers to substances that bind to a particular type of cells and enable the isolation of that cell from other cells.
  • exemplary capture agents are magnetic beads, ferrofluids, encapsulating reagents, molecules that bind the particular cell type and the like.
  • sample refers to a collection of similar fluids, cells, or tissues isolated from a subject, as well as fluids, cells, or tissues present within a subject.
  • exemplary samples are tissue biopsies, fine needle aspirations, surgically resected tissue, organ cultures, cell cultures and biological fluids such as blood, serum and serosal fluids, plasma, lymph, urine, saliva, cystic fluid, tear drops, feces, sputum, mucosal secretions of the secretory tissues and organs, vaginal secretions, ascites fluids, fluids of the pleural, pericardial, peritoneal, abdominal and other body cavities, fluids collected by bronchial lavage, synovial fluid, liquid solutions contacted with a subject or biological source, for example, cell and organ culture medium including cell or organ conditioned medium and lavage fluids and the like.
  • Substituting or “substituted” or “mutating” or “mutated” refers to altering, deleting of inserting one or more amino acids or nucleotides in a polypeptide or polynucleotide sequence to generate a variant of that sequence.
  • Variant refers to a polypeptide or a polynucleotide that differs from a reference polypeptide or a reference polynucleotide by one or more modifications for example, substitutions, insertions or deletions.
  • Specifically binds” or “specific binding” refers to the ability of the FN3 domain of the invention to bind PD-L1 with a dissociation constant (K D ) of about 1 ⁇ 10 ⁇ 6 M or less, for example about 1 ⁇ 10 ⁇ 7 M or less, about 1 ⁇ 10 ⁇ 8 M or less, about 1 ⁇ 10 ⁇ 9 M or less, about 1 ⁇ 10 ⁇ 10 M or less, about 1 ⁇ 10 ⁇ 11 M or less, about 1 ⁇ 10 ⁇ 12 M or less, or about 1 ⁇ 10 ⁇ 13 M or less.
  • K D dissociation constant
  • specific binding refers to the ability of the FN3 domain of the invention to bind PD-L1 at least 5-fold above the negative control in standard ELISA assay.
  • the isolated FN3 domain of the invention that specifically binds PD-L1 may, however, have cross-reactivity to other related antigens, for example to the same predetermined antigen from other species (homologs), such as Macaca Fascicularis (cynomolgous monkey, cyno) or Pan troglodytes (chimpanzee).
  • homologs such as Macaca Fascicularis (cynomolgous monkey, cyno) or Pan troglodytes (chimpanzee).
  • Library refers to a collection of variants.
  • the library may be composed of polypeptide or polynucleotide variants.
  • “Stability” refers to the ability of a molecule to maintain a folded state under physiological conditions such that it retains at least one of its normal functional activities, for example, binding to a predetermined antigen such as PD-L1.
  • PD-L1 refers to human PD-L1 protein having the amino acid sequence of SEQ ID NO: 32.
  • the extracellular domain of PD-L1 spans residues 1-220, the transmembrane domain spans residues 221-241 and the cytoplasmic domain spans residues 242-272.
  • PD-1 refers to human PD-1 protein having the amino acid sequence of SEQ ID NO: 33.
  • the extracellular domain of PD-1 spans residues 1-150, the transmembrane domain spans residues 151-171 and the cytoplasmic domain spans residues 172-268 of SEQ ID NO: 33.
  • Tencon refers to the synthetic fibronectin type III (FN3) domain having the sequence shown in SEQ ID NO: 1 and described in U.S. Pat. Publ. No. 2010/0216708.
  • a “cancer cell” or a “tumor cell” refers to a cancerous, pre-cancerous or transformed cell, either in vivo, ex vivo, and in tissue culture, that has spontaneous or induced phenotypic changes that do not necessarily involve the uptake of new genetic material.
  • transformation can arise from infection with a transforming virus and incorporation of new genomic nucleic acid, or uptake of exogenous nucleic acid, it can also arise spontaneously or following exposure to a carcinogen, thereby mutating an endogenous gene.
  • Transformation/cancer is exemplified by, e.g., morphological changes, immortalization of cells, aberrant growth control, foci formation, proliferation, malignancy, tumor specific markers levels, invasiveness, tumor growth or suppression in suitable animal hosts such as nude mice, and the like, in vitro, in vivo, and ex vivo (Freshney, Culture of Animal Cells: A Manual of Basic Technique (3rd ed. 1994)).
  • Vector refers to a polynucleotide capable of being duplicated within a biological system or that can be moved between such systems.
  • Vector polynucleotides typically contain elements, such as origins of replication, polyadenylation signal or selection markers that function to facilitate the duplication or maintenance of these polynucleotides in a biological system.
  • Examples of such biological systems may include a cell, virus, animal, plant, and reconstituted biological systems utilizing biological components capable of duplicating a vector.
  • the polynucleotide comprising a vector may be DNA or RNA molecules or a hybrid of these.
  • “Expression vector” refers to a vector that can be utilized in a biological system or in a reconstituted biological system to direct the translation of a polypeptide encoded by a polynucleotide sequence present in the expression vector.
  • Polynucleotide refers to a synthetic molecule comprising a chain of nucleotides covalently linked by a sugar-phosphate backbone or other equivalent covalent chemistry.
  • cDNA is a typical example of a polynucleotide.
  • Polypeptide or “protein” refers to a molecule that comprises at least two amino acid residues linked by a peptide bond to form a polypeptide. Small polypeptides of less than about 50 amino acids may be referred to as “peptides”.
  • Value refers to the presence of a specified number of binding sites specific for an antigen in a molecule.
  • the terms “monovalent”, “bivalent”, “tetravalent”, and “hexavalent” refer to the presence of one, two, four and six binding sites, respectively, specific for an antigen in a molecule.
  • Subject includes any human or nonhuman animal “Nonhuman animal” includes all vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dogs, cats, horses, cows chickens, amphibians, reptiles, etc. Except when noted, the terms “patient” or “subject” are used interchangeably.
  • Isolated refers to a homogenous population of molecules (such as synthetic polynucleotides or a polypeptide such as FN3 domains) which have been substantially separated and/or purified away from other components of the system the molecules are produced in, such as a recombinant cell, as well as a protein that has been subjected to at least one purification or isolation step.
  • molecules such as synthetic polynucleotides or a polypeptide such as FN3 domains
  • isolated FN3 domain refers to an FN3 domain that is substantially free of other cellular material and/or chemicals and encompasses FN3 domains that are isolated to a higher purity, such as to 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% purity.
  • the present invention provides fibronectin type III (FN3) domains that specifically bind PD-L1. These molecules can be used in therapeutic and diagnostic applications and in imaging.
  • the present invention provides polynucleotides encoding the FN3 domains of the invention or complementary nucleic acids thereof, vectors, host cells, and methods of making and using them.
  • the invention provides an isolated FN3 domain that specifically binds PD-L1.
  • the FN3 domain of the invention may bind PD-L1 with a dissociation constant (K D ) of less than about 1 ⁇ 10 ⁇ 7 M, for example less than about 1 ⁇ 10 ⁇ 8 M, less than about 1 ⁇ 10 ⁇ 9 M, less than about 1 ⁇ 10 ⁇ 1 ° M, less than about 1 ⁇ 10 ⁇ 11 M, less than about 1 ⁇ 10 ⁇ 12 M, or less than about 1 ⁇ 10 ⁇ 13 M as determined by surface plasmon resonance or the Kinexa method, as practiced by those of skill in the art.
  • K D dissociation constant
  • the measured affinity of a particular FN3 domain-antigen interaction can vary if measured under different conditions (e.g., osmolarity, pH).
  • affinity and other antigen-binding parameters e.g., K D , K on , K off
  • K D dissociation constant
  • the FN3 domain of the invention may bind PD-L1 at least 5-fold above the signal obtained for a negative control in standard ELISA assay.
  • the FN3 domain that specifically binds PD-L1 comprises an initiator methionine (Met) linked to the N-terminus of the molecule.
  • the FN3 domain that specifically binds PD-L1 comprises a cysteine (Cys) linked to a C-terminus of the FN3 domain.
  • N-terminal Met and/or the C-terminal Cys may facilitate expression and/or conjugation of half-life extending molecules.
  • the FN3 domain that specifically binds PD-L1 is internalized into a cell.
  • Internalization of the FN3 domain may facilitate delivery of a cytotoxic agent into tumor cells.
  • the FN3 domain that specifically binds PD-L1 inhibits binding of PD-L1 to PD-1.
  • Inhibition of binding of PD-L1 to PD-1 by the FN3 domains of the invention may be assessed using competition ELISA.
  • 1 ⁇ g/ml recombinant human PD-L1 extracellular domain is bound on wells of microtiter plates, the wells are washed and blocked, and 10 ⁇ g/ml of the test FN3 domain is added. Without washing, 7.5 ⁇ g/ml PD-1 extracellular domain is added into the wells and incubated for 30 min, after which 0.5 ⁇ g/ml anti-PD-1 antibody is added and incubated for 30 min.
  • the plates are washed and 0.5 ⁇ g/mL neutravidin-HRP conjugate polyclonal antibody is added and incubated for 30 minutes.
  • the plates are washed and POD Chemiluminescence substrate added immediately prior to reading the luminescence signal.
  • the FN3 domains of the invention inhibit binding of PD-L1 to PD-1 when the binding of PD-1 is reduced by at least about 80%, 85%, 90%, 95% or 100%.
  • the FN3 domain that specifically binds PD-L1 is a PD-L1 antagonist.
  • the FN3 domain that specifically binds PD-L1 is a PD-L1 agonist.
  • “Antagonist” refers to a FN3 domain that specifically binds PD-L1 that suppresses at least one reaction or activity that is induced by PD-L1 binding PD-1.
  • a molecule is an antagonist when the at least one reaction or activity is suppressed by at least about 30%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% more than the at least one reaction or activity suppressed in the absence of the antagonist (e.g., negative control), or when the suppression is statistically significant when compared to the suppression in the absence of the antagonist.
  • a typical reaction or activity that is induced by PD-L1 binding PD-1 is reduced antigen-specific CD4 + or CD8 + cell proliferation or reduced interferon- ⁇ (IFN- ⁇ ) production by T cells.
  • the antagonistic FN3 domains that specifically bind PD-L1 may be used in the treatment of cancer or viral infections and in general in treatment of diseases in which activation of immune responses is desirable.
  • “Agonist” refers to a FN3 domain that specifically binds PD-L1 that induces at least one reaction or activity that is induced by PD-L1 binding PD-1.
  • the FN3 domain is an agonist when the at least one reaction or activity is induced by at least about 30%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% greater than the at least one reaction or activity induced in the absence of the agonist (e.g., negative control), or when the induction is statistically significant when compared to the induction in the absence of the agonist.
  • a typical reaction or activity that is induced by PD-L1 binding PD-1 is reduced antigen-specific CD4 + or CD8 + cell proliferation or reduced interferon- ⁇ (IFN- ⁇ ) production by T cells.
  • the agonistic FN3 domains that specifically bind PD-L1 may be used in the treatment of autoimmune or inflammatory diseases and in general diseases in which suppression of immune responses is desirable.
  • the FN3 domain that specifically binds PD-L1 does not inhibit binding of PD-L1 to PD-1.
  • the FN3 domain that specifically binds PD-L1 does not activate signaling downstream of PD-1.
  • the FN3 domain that specifically binds PD-L1 is based on Tencon sequence of SEQ ID NO: 1 or Tencon 27 sequence of SEQ ID NO: 4, optionally having substitutions at residues positions 11, 14, 17, 37, 46, 73, and/or 86 (residue numbering corresponding to SEQ ID NO: 4).
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NOs: 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 34.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 35.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 36.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 37.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 38.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 39.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 40.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 41.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 42.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 43.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 44.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 45.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 46.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 47.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 48.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 49.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 50.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 51.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 52.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 53.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 54.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 55.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 56.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 57.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 58.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 59.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 60.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 61.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 62.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 63.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 64.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 65.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 66.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 67.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 68.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 69.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 70.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 71.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 72.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 73.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 74.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 75.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 76.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 77.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 78.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 79.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 80.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 81.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 82.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 83.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 84.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 85.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 86.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 87.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 88.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 89.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 90.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 91.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 92.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 93.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 94.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 95.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 96.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 97.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 98.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 99.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 100.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 101.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 102.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 103.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 104.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 105.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 106.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 107.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 108.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 109.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 110.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 111.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 112.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 113.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 114.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 115.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 116.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 117.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 118.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 119.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 120.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 121.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 122.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 123.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 124.
  • the isolated FN3 domain that specifically binds PD-L1 comprises an initiator methionine (Met) linked to the N-terminus of the molecule.
  • the isolated FN3 domain that specifically binds PD-L1 comprises an amino acid sequence that is 62%, 63%, 64% , 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO: 74.
  • the isolated FN3 domain that specifically binds PD-L1 comprises an amino acid sequence that is 62%, 63%, 64% , 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of any one of SEQ ID NOs: 34-124.
  • the invention also provides an isolated FN3 domain that specifically binds PD-L1 conjugated to a heterologous molecule(s).
  • the heterologous molecule is a detectable label or a cytotoxic agent.
  • the invention also provides an FN3 domain that specifically binds PD-L1 conjugated to a detectable label.
  • the invention also provides an FN3 domain that specifically binds PD-L1 conjugated to a cytotoxic agent.
  • the detectable label is also a cytotoxic agent.
  • the FN3 domains that specifically bind PD-L1 of the invention conjugated to a detectable label can be used to evaluate expression of PD-L1 on samples such as tumor tissue in vivo or in vitro.
  • Detectable label includes compositions that when conjugated to the FN3 domains that specifically bind PD-L 1 of the invention renders the latter detectable, via spectroscopic, photochemical, biochemical, immunochemical, or chemical means.
  • Exemplary detectable labels include radioactive isotopes, magnetic beads, metallic beads, colloidal particles, fluorescent dyes, electron-dense reagents, enzymes (for example, as commonly used in an ELISA), biotin, digoxigenin, haptens, luminescent molecules, chemiluminescent molecules, fluorochromes, fluorophores, fluorescent quenching agents, colored molecules, radioactive isotopes, scintillants, avidin, streptavidin, protein A, protein G, antibodies or fragments thereof, poly histidine, Ni2+, Flag tags, myc tags, heavy metals, enzymes, alkaline phosphatase, peroxidase, luciferase, electron donors/acceptors, acridinium esters, and colorimetric substrates.
  • enzymes for example, as commonly used in an ELISA
  • biotin digoxigenin
  • haptens luminescent molecules
  • chemiluminescent molecules chemiluminescent molecules
  • a detectable label may emit a signal spontaneously, such as when the detectable label is a radioactive isotope. In other cases the detectable label emits a signal as a result of being stimulated by an external field.
  • Exemplary radioactive isotopes may be y-emitting, Auger-emitting, ⁇ -emitting, an alpha-emitting or positron-emitting radioactive isotope.
  • Exemplary radioactive isotopes include 3 H, 11 C, 13 C, 15 N, 18 F, 19 F, 55 Co, 57 Co, 60 Co, 61 Cu, 62 Cu, 64 Cu, 67 Cu, 68 Cu, 72 As, 75 Br, 86 Y, 89 Zr, 90 Sr, 94m Tc, 99m Tc, 115 In, 123 I, 124 I, 125 I, 131 I, 211 At, 212 Bi, 213 Bi, 223 Ra, 226 Ra, 225 Ac and 227 Ac.
  • Exemplary metal atoms are metals with an atomic number greater than 20, such as calcium atoms, scandium atoms, titanium atoms, vanadium atoms, chromium atoms, manganese atoms, iron atoms, cobalt atoms, nickel atoms, copper atoms, zinc atoms, gallium atoms, germanium atoms, arsenic atoms, selenium atoms, bromine atoms, krypton atoms, rubidium atoms, strontium atoms, yttrium atoms, zirconium atoms, niobium atoms, molybdenum atoms, technetium atoms, ruthenium atoms, rhodium atoms, palladium atoms, silver atoms, cadmium atoms, indium atoms, tin atoms, antimony atoms, tellurium atoms, iodine atoms,
  • the metal atoms may be alkaline earth metals with an atomic number greater than twenty.
  • the metal atoms may be lanthanides.
  • the metal atoms may be actinides.
  • the metal atoms may be transition metals.
  • the metal atoms may be poor metals.
  • the metal atoms may be gold atoms, bismuth atoms, tantalum atoms, and gadolinium atoms.
  • the metal atoms may be metals with an atomic number of 53 (i.e. iodine) to 83 (i.e. bismuth).
  • the metal atoms may be atoms suitable for magnetic resonance imaging.
  • the metal atoms may be metal ions in the form of +1 , +2, or +3 oxidation states, such as Ba 2+ , Bi 3+ , Cs + , Ca 2+ , Cr 2+ , Cr 3+ , Cr 6+ , Co 3+ , Co 3+ , Cu + , Cu 2+ , Cu 3+ , Ga 3+ , Gd 3+ , Au + , Au 3+ , Pe 2+ , Fe 3+ , F 3+ , Pb 2+ , Mn 2+ , Mn 3+ , Mn 4+ , Mn 7+ , Hg 2+ , Ni 2+ , Ni 3+ , Ag + , Sr 2+ , Sn 2+ , Sn 4+ , and Zn 2+ .
  • the metal atoms may comprise a metal oxide, such as iron oxide, manganese oxide, or gadolinium oxide.
  • Suitable dyes include any commercially available dyes such as, for example, 5(6)-carboxyfluorescein, IRDye 680RD maleimide or IRDye 800CW, ruthenium polypyridyl dyes, and the like.
  • Suitable fluorophores are fluorescein isothiocyante (FITC), fluorescein thiosemicarbazide, rhodamine, Texas Red, CyDyes (e.g., Cy3, Cy5, Cy5.5), Alexa Fluors (e.g., Alexa488, Alexa555, Alexa594; Alexa647), near infrared (NIR) (700-900 nm) fluorescent dyes, and carbocyanine and aminostyryl dyes.
  • FITC fluorescein isothiocyante
  • fluorescein thiosemicarbazide e.g., Texas Red
  • CyDyes e.g., Cy3, Cy5, Cy5.5
  • Alexa Fluors e.g., Alexa488, Alexa555, Alexa594; Alexa647
  • NIR near infrared
  • the FN3 domains that specifically bind PD-L1 conjugated to a detectable label may be used as an imaging agent to evaluate tumor distribution, diagnosis for the presence of tumor cells and/or, recurrence of tumor.
  • the FN3 domains that specifically bind PD-L1 of the invention are conjugated to a cytotoxic agent.
  • the cytotoxic agent is a chemotherapeutic agent, a drug, a growth inhibitory agent, a toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
  • a chemotherapeutic agent e.g., a drug, a growth inhibitory agent, a toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
  • the FN3 domains that specifically bind PD-L1 conjugated to a cytotoxic agent of the invention may be used in the targeted delivery of the cytotoxic agent to PD-L1 expressing tumor cell, and intracellular accumulation therein, wherein systemic administration of these unconjugated cytotoxic agents may result in unacceptable levels of toxicity to normal cells.
  • the cytotoxic agent is daunomycin, doxorubicin, methotrexate, vindesine, bacterial toxins such as diphtheria toxin, ricin, geldanamycin, maytansinoids or calicheamicin.
  • the cytotoxic agent may elict their cytotoxic and cytostatic effects by mechanisms including tubulin binding, DNA binding, or topoisomerase inhibition.
  • the cytotoxic agent is an enzymatically active toxins such as diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa ), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes.
  • enzymatically active toxins such as diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa ), ricin A chain,
  • the cytotoxic agent is a radionuclide, such as 212 Bi, 131 I, 131 In, 90 Y, and 186 Re.
  • the cytotoxic agent is dolastatins or dolostatin peptidic analogs and derivatives, auristatin or monomethyl auristatin phenylalanine
  • exemplary molecules are disclosed in U.S. Pat Nos. 5,635,483 and 5,780,588. Dolastatins and auristatins have been shown to interfere with microtubule dynamics, GTP hydrolysis, and nuclear and cellular division (Woyke et al (2001) Antimicrob Agents and Chemother. 45(12):3580-3584) and have anticancerand antifungal activity.
  • the dolastatin or auristatin drug moiety may be attached to the FN3 domain of the invention through the N (amino) terminus or the C (carboxyl) terminus of the peptidic drug moiety (WO 02/088172), or via any cysteine engineered into the FN3 domain.
  • the FN3 domains that specifically bind PD-L1 of the invention may be conjugated to a detectable label using known methods.
  • the detectable label is complexed with a chelating agent.
  • the detectable label is conjugated to the FN3 domain that specifically binds PD-L1 of the invention via a linker.
  • the detectable label or the cytotoxic moiety may be linked directly, or indirectly, to the FN3 domain that specifically binds PD-L1 of the invention using known methods.
  • Suitable linkers are known in the art and include, for example, prosthetic groups, non-phenolic linkers (derivatives of N-succimidyl-benzoates; dodecaborate), chelating moieties of both macrocyclics and acyclic chelators, such as derivatives of 1,4,7,10-tetraazacyclododecane-1,4,7,10,tetraacetic acid (DOTA), derivatives of diethylenetriaminepentaacetic avid (DTPA), derivatives of S-2-(4-Isothiocyanatobenzyl)-1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA) and derivatives of 1,4,8,11-tetraazacy clodocedan-1,4,8,11-tetraacetic acid (TETA), N
  • the FN3 domain that specifically binds PD-L1 is removed from the blood via renal clearance.
  • Tencon is a non-naturally occurring fibronectin type III (FN3) domain designed from a consensus sequence of fifteen FN3 domains from human tenascin-C (Jacobs et al., Protein Engineering, Design, and Selection, 25:107-117, 2012; U.S. Pat. Publ. No. 2010/0216708).
  • the crystal structure of Tencon shows six surface-exposed loops that connect seven beta-strands as is characteristic to the FN3 domains, the beta-strands referred to as A, B, C, D, E, F, and G, and the loops referred to as AB, BC, CD, DE, EF, and FG loops (Bork and Doolittle, Proc Natl Acad Sci USA 89:8990-8992, 1992; U.S. Pat. No. 6,673,901). These loops, or selected residues within each loop, may be randomized in order to construct libraries of fibronectin type III (FN3) domains that may be used to select novel molecules that bind Pd-L1. Table 1 shows positions and sequences of each loop and beta-strand in Tencon (SEQ ID NO: 1).
  • Library designed based on Tencon sequence may thus have randomized FG loop, or randomized BC and FG loops, such as libraries TCL1 or TCL2 as described below.
  • the Tencon BC loop is 7 amino acids long, thus 1, 2, 3, 4, 5, 6 or 7 amino acids may be randomized in the library diversified at the BC loop and designed based on Tencon sequence.
  • the Tencon FG loop is 7 amino acids long, thus 1, 2, 3, 4, 5, 6 or 7 amino acids may be randomized in the library diversified at the FG loop and designed based on Tencon sequence.
  • Further diversity at loops in the Tencon libraries may be achieved by insertion and/or deletions of residues at loops.
  • the FG and/or BC loops may be extended by 1-22 amino acids, or decreased by 1-3 amino acids.
  • the FG loop in Tencon is 7 amino acids long, whereas the corresponding loop in antibody heavy chains ranges from 4-28 residues.
  • the FG loop may be diversified in sequence as well as in length to correspond to the antibody CDR3 length range of 4-28 residues.
  • the FG loop can further be diversified in length by extending the loop by additional 1, 2, 3, 4 or 5 amino acids.
  • Library designed based on Tencon sequence may also have randomized alternative surfaces that form on a side of the FN3 domain and comprise two or more beta strands, and at least one loop.
  • One such alternative surface is formed by amino acids in the C and the F beta-strands and the CD and the FG loops (a C-CD-F-FG surface).
  • a library design based on Tencon alternative C-CD-F-FG surface is described in U.S. Pat. Publ. No. US2013/0226834.
  • Library designed based on Tencon sequence also includes libraries designed based on Tencon variants, such as Tencon variants having substitutions at residues positions 11, 14, 17, 37, 46, 73, or 86 (residue numbering corresponding to SEQ ID NO: 1), and which variants display improve thermal stability.
  • Exemplary Tencon variants are described in US Pat. Publ. No. 2011/0274623, and include Tencon27 (SEQ ID NO: 4) having substitutions E 11R, L17A, N46V and E861 when compared to Tencon of SEQ ID NO: 1.
  • Tencon and other FN3 sequence based libraries may be randomized at chosen residue positions using a random or defined set of amino acids.
  • variants in the library having random substitutions may be generated using NNK codons, which encode all 20 naturally occurring amino acids.
  • DVK codons may be used to encode amino acids Ala, Trp, Tyr, Lys, Thr, Asn, Lys, Ser, Arg, Asp, Glu, Gly, and Cys.
  • NNS codons may be used to give rise to all 20 amino acid residues and simultaneously reducing the frequency of stop codons.
  • FN3 domains with biased amino acid distribution at positions to be diversified may be synthesized for example using Slonomics® technology (http:_/www_sloning_com). This technology uses a library of pre-made double stranded triplets that act as universal building blocks sufficient for thousands of gene synthesis processes.
  • the triplet library represents all possible sequence combinations necessary to build any desired DNA molecule.
  • the codon designations are according to the well-known IUB code.
  • the FN3 domains that specifically bind PD-L1 of the invention may be isolated by producing the FN3 library such as the Tencon library using cis display to ligate DNA fragments encoding the scaffold proteins to a DNA fragment encoding RepA to generate a pool of protein-DNA complexes formed after in vitro translation wherein each protein is stably associated with the DNA that encodes it (U.S. Pat. No. 7,842,476; Odegrip et al., Proc Natl Acad Sci U S A 101, 2806-2810, 2004), and assaying the library for specific binding to PSMA by any method known in the art and described in the Example.
  • the FN3 library such as the Tencon library using cis display to ligate DNA fragments encoding the scaffold proteins to a DNA fragment encoding RepA to generate a pool of protein-DNA complexes formed after in vitro translation wherein each protein is stably associated with the DNA that encodes it (U.S. Pat. No. 7,842,
  • exemplary well known methods which can be used are ELISA, sandwich immunoassays, and competitive and non-competitive assays (see, e.g., Ausubel et al., eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York).
  • the identified FN3 domains that specifically bind PD-L1 are further characterized for their binding to PD-L1, modulation of PD-L1 activity, internalization, stability, and other desired characteristics.
  • the FN3 domains that specifically bind PD-L1 of the invention may be generated using any FN3 domain as a template to generate a library and screening the library for molecules specifically binding PD-L1 using methods provided within.
  • Exemplar FN3 domains that may be used are the 3rd FN3 domain of tenascin C (TN3) (SEQ ID NO: 125), Fibcon (SEQ ID NO: 126), and the 10 th FN3 domain of fibronectin (FN10) (SEQ ID NO: 127).
  • Standard cloning and expression techniques are used to clone the libraries into a vector or synthesize double stranded cDNA cassettes of the library, to express, or to translate the libraries in vitro.
  • ribosome display Hanes and Pluckthun, Proc Natl Acad Sci USA, 94, 4937-4942, 1997)
  • mRNA display Robots and Szostak, Proc Natl Acad Sci USA, 94, 12297-12302, 1997)
  • other cell-free systems U.S. Pat. No. 5,643,768
  • the libraries of the FN3 domain variants may be expressed as fusion proteins displayed on the surface for example of any suitable bacteriophage. Methods for displaying fusion polypeptides on the surface of a bacteriophage are well known (U. S. Pat. Publ. No. 2011/0118144; Int. Pat. Publ. No.
  • the FN3 domain that specifically binds PD-L1 is based on Tencon sequence of SEQ ID NO: 1 or Tencon27 sequence of SEQ ID NO: 4, the SEQ ID NO: 1 or the SEQ ID NO: 4, optionally having substitutions at residues positions 11, 14, 17, 37, 46, 73, and/or 86.
  • the FN3 domains that specifically bind PD-L1 of the invention may be modified to improve their properties such as improve thermal stability and reversibility of thermal folding and unfolding.
  • Several methods have been applied to increase the apparent thermal stability of proteins and enzymes, including rational design based on comparison to highly similar thermostable sequences, design of stabilizing disulfide bridges, mutations to increase alpha-helix propensity, engineering of salt bridges, alteration of the surface charge of the protein, directed evolution, and composition of consensus sequences (Lehmann and Wyss, Curr Opin Biotechnol, 12, 371-375, 2001).
  • High thermal stability may increase the yield of the expressed protein, improve solubility or activity, decrease immunogenicity, and minimize the need of a cold chain in manufacturing.
  • Residues that may be substituted to improve thermal stability of Tencon are residue positions 11, 14, 17, 37, 46, 73, or 86, and are described in US Pat. Publ. No. 2011/0274623. Substitutions corresponding to these residues may be incorporated to the FN3 domain containing molecules of the invention.
  • Proteins are sensitive or “labile” to denaturation caused by heat, by ultraviolet or ionizing radiation, changes in the ambient osmolarity and pH if in liquid solution, mechanical shear force imposed by small pore-size filtration, ultraviolet radiation, ionizing radiation, such as by gamma irradiation, chemical or heat dehydration, or any other action or force that may cause protein structure disruption.
  • the stability of the molecule can be determined using standard methods. For example, the stability of a molecule can be determined by measuring the thermal melting (“T m ”) temperature, the temperature in ° Celsius (° C.) at which half of the molecules become unfolded, using standard methods. Typically, the higher the T m , the more stable the molecule.
  • T m thermal melting
  • the chemical environment also changes the ability of the protein to maintain a particular three dimensional structure.
  • the FN3 domain that specifically binds PD-L1 of the invention may exhibit increased stability by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% or more compared to the same domain prior to engineering measured by the increase in the T..
  • Chemical denaturation can likewise be measured by a variety of methods.
  • Chemical denaturants include guanidinium hydrochloride, guanidinium thiocyanate, urea, acetone, organic solvents (DMF, benzene, acetonitrile), salts (ammonium sulfate, lithium bromide, lithium chloride, sodium bromide, calcium chloride, sodium chloride); reducing agents (e.g. dithiothreitol, beta-mercaptoethanol, dinitrothiobenzene, and hydrides, such as sodium borohydride), non-ionic and ionic detergents, acids (e.g.
  • hydrochloric acid HCl
  • acetic acid CH 3 COOH
  • halogenated acetic acids hydrophobic molecules
  • targeted denaturants e.g. phosopholipids
  • Quantitation of the extent of denaturation can rely on loss of a functional property, such as ability to bind a target molecule, or by physiochemical properties, such as tendency to aggregation, exposure of formerly solvent inaccessible residues, or disruption or formation of disulfide bonds.
  • the FN3 domain that specifically binds PD-L1 of the invention may be generated as monomers, dimers, or multimers, for example, as a means to increase the valency and thus the avidity of target molecule binding, or to generate bi- or multispecific scaffolds simultaneously binding two or more different target molecules.
  • the dimers and multimers may be generated by linking monospecific, bi- or multispecific protein scaffolds, for example, by the inclusion of an amino acid linker, for example a linker containing poly-glycine, glycine and serine, or alanine and proline.
  • Exemplary linker include (GS)2, (SEQ ID NO: 128), (GGGS)2 (SEQ ID NO: 129), (GGGGS)5 (SEQ ID NO: 130), (AP) 2 (SEQ ID NO: 131), (AP) 5 (SEQ ID NO: 132), (AP) 10 (SEQ ID NO: 133), (AP) 20 (SEQ ID NO: 134) and A(EAAAK) 5 AAA (SEQ ID NO: 135).
  • the dimers and multimers may be linked to each other in a N-to C-direction.
  • the FN3 domains that specifically bind PD-L1 of the invention may incorporate other subunits for example via covalent interaction.
  • the FN3 domains that specifically bind PD-L1 of the invention further comprise a half-life extending moiety.
  • Exemplary half-life extending moieties are albumin, albumin variants, albumin-binding proteins and/or domains, transferrin and fragments and analogues thereof, and Fc regions.
  • An exemplary albumin variant is shown in SEQ ID NO: 136. Amino acid sequences of the human Fc regions are well known, and include IgG1, IgG2, IgG3, IgG4, IgM, IgA and IgE Fc regions.
  • All or a portion of an antibody constant region may be attached to the FN3 domain that specifically binds PD-L1 of the invention to impart antibody-like properties, especially those properties associated with the Fc region, such as Fc effector functions such as Clq binding, complement dependent cytotoxicity (CDC), Fc receptor binding, antibody-dependent cell-mediated cytotoxicity (ADCC), phagocytosis, down regulation of cell surface receptors (e.g., B cell receptor; BCR), and may be further modified by modifying residues in the Fc responsible for these activities (for review; see Strohl, Curr Opin Biotechnol. 20, 685-691, 2009).
  • Fc effector functions such as Clq binding, complement dependent cytotoxicity (CDC), Fc receptor binding, antibody-dependent cell-mediated cytotoxicity (ADCC), phagocytosis, down regulation of cell surface receptors (e.g., B cell receptor; BCR)
  • CDC complement dependent cytotoxicity
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • Additional moieties may be incorporated into the FN3 domains that specifically bind PD-L1 of the invention such as polyethylene glycol (PEG) molecules, such as PEG5000 or PEG20,000, fatty acids and fatty acid esters of different chain lengths, for example laurate, myristate, stearate, arachidate, behenate, oleate, arachidonate, octanedioic acid, tetradecanedioic acid, octadecanedioic acid, docosanedioic acid, and the like, polylysine, octane, carbohydrates (dextran, cellulose, oligo- or polysaccharides) for desired properties.
  • PEG polyethylene glycol
  • fatty acids and fatty acid esters of different chain lengths for example laurate, myristate, stearate, arachidate, behenate, oleate, arachidonate, octanedio
  • a pegyl moiety may for example be added to the FN3 domain that specifically binds PD-L1 of the invention by incorporating a cysteine residue to the C-terminus of the molecule, or engineering cysteines into residue positions that face away from the PD-L1 binding face of the molecule, and attaching a pegyl group to the cysteine using well known methods.
  • FN3 domains that specifically bind PD-L1 of the invention incorporating additional moieties may be compared for functionality by several well-known assays.
  • altered properties due to incorporation of Fc domains and/or Fc domain variants may be assayed in Fc receptor binding assays using soluble forms of the receptors, such as the Fc ⁇ RI, Fc ⁇ RII, Fc ⁇ RIII or FcRn receptors, or using well known cell-based assays measuring for example ADCC or CDC, or evaluating pharmacokinetic properties of the molecules of the invention in in vivo models.
  • the invention also provides nucleic acids encoding the FN3 domains specifically binding PD-L1 of the invention as isolated polynucleotides or as portions of expression vectors or as portions of linear DNA sequences, including linear DNA sequences used for in vitro transcription/translation, vectors compatible with prokaryotic, eukaryotic or filamentous phage expression, secretion and/or display of the compositions or directed mutagens thereof.
  • Certain exemplary polynucleotides are disclosed herein, however, other polynucleotides which, given the degeneracy of the genetic code or codon preferences in a given expression system, encode the FN3 domains of the invention are also within the scope of the invention.
  • the invention also provides an isolated polynucleotide encoding the FN3 domain specifically binding PD-L1 comprising the amino acid sequence of SEQ ID NOs: 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117
  • the polynucleotides of the invention may be produced by chemical synthesis such as solid phase polynucleotide synthesis on an automated polynucleotide synthesizer and assembled into complete single or double stranded molecules.
  • the polynucleotides of the invention may be produced by other techniques such as PCR followed by routine cloning. Techniques for producing or obtaining polynucleotides of a given known sequence are well known in the art.
  • the polynucleotides of the invention may comprise at least one non-coding sequence, such as a promoter or enhancer sequence, intron, polyadenylation signal, a cis sequence facilitating RepA binding, and the like.
  • the polynucleotide sequences may also comprise additional sequences encoding additional amino acids that encode for example a marker or a tag sequence such as a histidine tag or an HA tag to facilitate purification or detection of the protein, a signal sequence, a fusion protein partner such as RepA, Fc or bacteriophage coat protein such as pIX or pIII.
  • the invention also provides a vector comprising at least one polynucleotide of the invention.
  • vectors may be plasmid vectors, viral vectors, vectors for baculovirus expression, transposon based vectors or any other vector suitable for introduction of the polynucleotides of the invention into a given organism or genetic background by any means.
  • vectors may be expression vectors comprising nucleic acid sequence elements that can control, regulate, cause or permit expression of a polypeptide encoded by such a vector.
  • Such elements may comprise transcriptional enhancer binding sites, RNA polymerase initiation sites, ribosome binding sites, and other sites that facilitate the expression of encoded polypeptides in a given expression system.
  • Such expression systems may be cell-based, or cell-free systems well known in the art.
  • the invention also provides a host cell comprising the vector of the invention.
  • the FN3 domain that specifically bind PD-L1 of the invention may be optionally produced by a cell line, a mixed cell line, an immortalized cell or clonal population of immortalized cells, as well known in the art. See, e.g., Ausubel, et al., ed., Current Protocols in Molecular Biology, John Wiley & Sons, Inc., NY, N.Y. (1987-2001); Sambrook, et al., Molecular Cloning: A Laboratory Manual, r d Edition, Cold Spring Harbor, N.Y. (1989); Harlow and Lane, Antibodies, a Laboratory Manual, Cold Spring Harbor, N.Y.
  • the host cell chosen for expression may be of mammalian origin or may be selected from COS-1, COS-7, HEK293, BHK21, CHO, BSC-1, He G2, SP2/0, HeLa, myeloma, lymphoma, yeast, insect or plant cells, or any derivative, immortalized or transformed cell thereof.
  • the host cell may be selected from a species or organism incapable of glycosylating polypeptides, e.g. a prokaryotic cell or organism, such as BL21, BL21(DE3), BL21-GOLD(DE3), XL1-Blue, JM109, HMS174, HMS174(DE3), and any of the natural or engineered E. coli spp, Klebsiella spp., or Pseudomonas spp strains.
  • the invention also provides a method of producing the isolated FN3 domain that specifically binds PD-L1 of the invention, comprising culturing the isolated host cell of the invention under conditions such that the isolated FN3 domain that specifically binds PD-L1 is expressed, and purifying the FN3 domain.
  • the FN3 domains that specifically bind PD-L1 may be purified from recombinant cell cultures by well-known methods, for example by protein A purification, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography, or high performance liquid chromatography (HPLC).
  • protein A purification ammonium sulfate or ethanol precipitation
  • acid extraction anion or cation exchange chromatography
  • phosphocellulose chromatography phosphocellulose chromatography
  • hydrophobic interaction chromatography affinity chromatography
  • hydroxylapatite chromatography and lectin chromatography or high performance liquid chromatography (HPLC).
  • the present invention also provides an anti-idiotypic antibody binding to the FN3 domain of the invention.
  • the invention also provides an anti-idiotypic antibody that specifically binds the FN3 domain comprising any one of SEQ ID NOs: 34-124.
  • the invention also provides a kit comprising the FN3 domain that specifically binds PD-L1 of the invention.
  • the kit may be used for therapeutic uses and as a diagnostic kit.
  • the kit comprises the FN3 domain that specifically binds PD-L1 of the invention and reagents for detecting the FN3 domain.
  • the kit can include one or more other elements including: instructions for use; other reagents, e.g., a label, an agent useful for chelating, or otherwise coupling, a radioprotective composition; devices or other materials for preparing the FN3 domain that specifically binds PD-L1 of the invention for administration for imaging, diagnostic or therapeutic purpose; pharmaceutically acceptable carriers; and devices or other materials for administration to a subject.
  • the kit comprises the FN3 domain that specifically binds PD-L1 comprising any one of SEQ ID NOs: 34-124.
  • the FN3 domains that specifically bind PD-L1 of the invention may be used to diagnose, monitor, modulate, treat, alleviate, help prevent the incidence of, or reduce the symptoms of human disease or specific pathologies in cells, tissues, organs, fluid, or, generally, a host.
  • the FN3 domains that specifically bind PD-L1 of the invention may also be used in imaging PD-L1 positive tumor tissue in a subject.
  • the methods of the invention may be used with an animal patient belonging to any classification. Examples of such animals include mammals such as humans, rodents, dogs, cats and farm animals.
  • the invention provides a method of diagnosing a subject having, or who is likely to develop cancer of a tissue based on the expression of PD-L1 by cells of the cancer tissue, methods of predicting success of immunotherapy, methods of prognosis, and methods of treatment.
  • the invention also provides a method of detecting PD-L1-expressing cancer cells in a tumor tissue, comprising
  • detecting whether PD-L1 is expressed in the tumor tissue by contacting toe sample of the tumor tissues with the FN3 domain that specifically binds PD-L1 comprising the sequence of any one of SEQ ID NOs: 34-124 and detecting the binding between PD-L1 and the FN3 domain.
  • the tissue can be tissue of any organ or anatomical system, for example lung, epithelial, connective, vascular, muscle, neural, skeletal, lymphatic, prostate, cervical, breast, spleen, gastric, intestinal, oral, esophageal, uterine, ovarian, renal or testicular tissue.
  • organ or anatomical system for example lung, epithelial, connective, vascular, muscle, neural, skeletal, lymphatic, prostate, cervical, breast, spleen, gastric, intestinal, oral, esophageal, uterine, ovarian, renal or testicular tissue.
  • PD-L1 expression may be evaluated using known methods such as immunohistochemistry or ELISA.
  • the invention also provides a method of isolating PD-L1 expressing cells, comprising
  • the invention also provides a method of detecting PD-L1-expressing cancer cells in a tumor tissue, comprising
  • the invention also provides a method of treating a subject having cancer, comprising administering to the subject a FN3 domain that specifically binds PD-L1 of the invention.
  • the subject has a solid tumor.
  • the subject has a hematological malignancy.
  • the solid tumor is a melanoma.
  • the solid tumor is a lung cancer.
  • the solid tumor is a non-small cell lung cancer (NSCLC).
  • NSCLC non-small cell lung cancer
  • the solid tumor is a squamous non-small cell lung cancer (NSCLC).
  • NSCLC squamous non-small cell lung cancer
  • the solid tumor is a non-squamous NSCLC.
  • the solid tumor is a lung adenocarcinoma.
  • the solid tumor is a renal cell carcinoma (RCC).
  • RRC renal cell carcinoma
  • the solid tumor is a mesothelioma.
  • the solid tumor is a nasopharyngeal carcinoma (NPC).
  • NPC nasopharyngeal carcinoma
  • the solid tumor is a colorectal cancer.
  • the solid tumor is a prostate cancer.
  • the solid tumor is castration-resistant prostate cancer.
  • the solid tumor is a stomach cancer.
  • the solid tumor is an ovarian cancer.
  • the solid tumor is a gastric cancer.
  • the solid tumor is a liver cancer.
  • the solid tumor is pancreatic cancer.
  • the solid tumor is a thyroid cancer.
  • the solid tumor is a squamous cell carcinoma of the head and neck.
  • the solid tumor is a carcinomas of the esophagus or gastrointestinal tract.
  • the solid tumor is a breast cancer.
  • the solid tumor is a fallopian tube cancer.
  • the solid tumor is a brain cancer.
  • the solid tumor is an urethral cancer.
  • the solid tumor is a genitourinary cancer.
  • the solid tumor is an endometriosis.
  • the solid tumor is a cervical cancer.
  • the solid tumor is a metastatic lesion of the cancer.
  • the hematological malignancy is a lymphoma, a myeloma or a leukemia.
  • the hematological malignancy is a B cell lymphoma.
  • the hematological malignancy is Burkitt's lymphoma.
  • the hematological malignancy is Hodgkin's lymphoma.
  • the hematological malignancy is a non-Hodgkin's lymphoma.
  • the hematological malignancy is a myelodysplastic syndrome.
  • the hematological malignancy is an acute myeloid leukemia (AML).
  • AML acute myeloid leukemia
  • the hematological malignancy is a chronic myeloid leukemia (CML).
  • CML chronic myeloid leukemia
  • the hematological malignancy is a chronic myelomoncytic leukemia (CMML).
  • CMML chronic myelomoncytic leukemia
  • the hematological malignancy is a multiple myeloma (MM).
  • the hematological malignancy is a plasmacytoma.
  • the cancer is kidney cancer.
  • Treating” or “treatment” refers to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) an undesired physiological change or disorder, such as the development or spread of cancer.
  • beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented.
  • a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve a desired therapeutic result.
  • a therapeutically effective amount of the FN3 domains that specifically bind PD-L1 of the invention may vary according to factors such as the disease state, age, sex, and weight of the individual. Exemplary indicators of an effective FN3 domain that specifically binds PD-L1 is improved well-being of the patient, decrease or shrinkage of the size of a tumor, arrested or slowed growth of a tumor, and/or absence of metastasis of cancer cells to other locations in the body.
  • the invention provides for pharmaceutical compositions of the FN3 domains that specifically bind PD-L1, optionally conjugated to a detectable label or a cytotoxic drug of the invention and a pharmaceutically acceptable carrier.
  • the FN3 domains that specifically bind PD-L1 of the invention may be prepared as pharmaceutical compositions containing an effective amount of the domain or molecule as an active ingredient in a pharmaceutically acceptable carrier.
  • Carrier refers to a diluent, adjuvant, excipient, or vehicle with which the active compound is administered.
  • Such vehicles can be liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
  • compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, stabilizing, thickening, lubricating and coloring agents, etc.
  • concentration of the molecules of the invention in such pharmaceutical formulation can vary widely, i.e., from less than about 0.5%, usually at least about 1% to as much as 15 or 20% by weight and will be selected primarily based on required dose, fluid volumes, viscosities, etc., according to the particular mode of administration selected.
  • Suitable vehicles and formulations, inclusive of other human proteins, e.g., human serum albumin, are described, for example, in e.g. Remington: The Science and Practice of Pharmacy, 21′ Edition, Troy, D.B. ed., Lipincott Williams and Wilkins, Philadelphia, PA 2006, Part 5, Pharmaceutical Manufacturing pp 691-1092, See especially pp. 958-989.
  • the mode of administration for therapeutic use of the FN3 domains of the invention may be any suitable route that delivers the agent to the host, such as parenteral administration, e.g., intradermal, intramuscular, intraperitoneal, intravenous or subcutaneous, pulmonary; transmucosal (oral, intranasal, intravaginal, rectal), using a formulation in a tablet, capsule, solution, powder, gel, particle; and contained in a syringe, an implanted device, osmotic pump, cartridge, micropump; or other means appreciated by the skilled artisan, as well known in the art.
  • parenteral administration e.g., intradermal, intramuscular, intraperitoneal, intravenous or subcutaneous, pulmonary
  • transmucosal oral, intranasal, intravaginal, rectal
  • a formulation in a tablet, capsule, solution, powder, gel, particle and contained in a syringe
  • an implanted device osmotic pump
  • Site specific administration may be achieved by for example intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracerebellar, intracerebroventricular, intracolic, intracervical, intragastric, intrahepatic, intracardial, intraosteal, intrapelvic, intrapericardiac, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravascular, intravesical, intralesional, vaginal, rectal, buccal, sublingual, intranasal, or transdermal delivery.
  • compositions can be supplied as a kit comprising a container that comprises the pharmaceutical composition as described herein.
  • a pharmaceutical composition can be provided, for example, in the form of an injectable solution for single or multiple doses, or as a sterile powder that will be reconstituted before injection.
  • a kit can include a dry-powder disperser, liquid aerosol generator, or nebulizer for administration of a pharmaceutical composition.
  • Such a kit can further comprise written information on indications and usage of the pharmaceutical composition.
  • Tencon is an immunoglobulin-like scaffold, fibronectin type III (FN3) domain, designed from a consensus sequence of fifteen FN3 domains from human tenascin-C (Jacobs et al., Protein Engineering, Design, and Selection, 25:107-117, 2012; U.S. Pat. No. 8,278,419).
  • the crystal structure of Tencon shows six surface-exposed loops that connect seven beta-strands. These loops, or selected residues within each loop, can be randomized in order to construct libraries of fibronectin type III (FN3) domains that can be used to select novel molecules that bind to specific targets.
  • a library designed to randomize only the FG loop of Tencon (SEQ ID NO: 1), TCL1, was constructed for use with the cis-display system (Jacobs et al., Protein Engineering, Design, and Selection, 25:107-117, 2012).
  • a single-strand DNA incorporating sequences for a Tac promoter, Tencon library coding sequence, RepA coding sequence, cis-element, and ori element is produced.
  • a complex is produced of the Tencon-RepA fusion protein bound in cis to the DNA from which it is encoded. Complexes that bind to a target molecule are then isolated and amplified by polymerase chain reaction (PCR), as described below.
  • TCL1 library for use with cis-display was achieved by successive rounds of PCR to produce the final linear, double-stranded DNA molecules in two halves; the 5′ fragment contains the promoter and Tencon sequences, while the 3′ fragment contains the repA gene and the cis- and ori elements. These two halves are combined by restriction digest in order to produce the entire construct.
  • the TCL1 library was designed to incorporate random amino acids only in the FG loop of Tencon, KGGHRSN (SEQ ID NO: 55). NNS codons were used in the construction of this library, resulting in the possible incorporation of all 20 amino acids and one stop codon into the FG loop.
  • the TCL1 library contains six separate sub-libraries, each having a different randomized FG loop length, from 7 to 12 residues, in order to further increase diversity.
  • TCL1 library (SEQ ID NO: 2) LPAPKNLVVSEVTEDSLRLSWTAPDAAFDSFLIQYQESEKVGEAINLTVP GSERSYDLTGLKPGTEYTVSIYGVX 1 X 2 X 3 X 4 X 5 X 6 X 7 X 8 X 9 X 10 X 11 X 12 PLSAEFTT; wherein X 1 , X 2 , X 3 , X 4 , X 5 , X 6 , X 7 is any amino acid; and X 8 , X 9 , X 10 , X 11 and X 12 are any amino acid or deleted Construction of TCL2 Library
  • TCL2 library was constructed in which both the BC and the FG loops of Tencon were randomized and the distribution of amino acids at each position was strictly controlled.
  • Table 3 shows the amino acid distribution at desired loop positions in the TCL2 library.
  • the designed amino acid distribution had two aims.
  • the library was biased toward residues that were predicted to be structurally important for Tencon folding and stability based on analysis of the Tencon crystal structure and/or from homology modeling. For example, position 29 was fixed to be only a subset of hydrophobic amino acids, as this residue was buried in the hydrophobic core of the Tencon fold.
  • a second layer of design included biasing the amino acid distribution toward that of residues preferentially found in the heavy chain HCDR3 of antibodies, to efficiently produce high-affinity binders (Birtalan et al., J Mol Biol 377:1518-28, 2008; Olson et al., Protein Sci 16:476-84, 2007).
  • the “designed distribution” in Table 2 refers to the distribution as follows: 6% alanine, 6% arginine, 3.9% asparagine, 7.5% aspartic acid, 2.5% glutamic acid, 1.5% glutamine, 15% glycine, 2.3% histidine, 2.5% isoleucine, 5% leucine, 1.5% lysine, 2.5% phenylalanine, 4% proline, 10% serine, 4.5% threonine, 4% tryptophan, 17.3% tyrosine, and 4% valine.
  • This distribution is devoid of methionine, cysteine, and STOP codons.
  • TCL2 library (SEQ ID NO: 3) LPAPKNLVVSEVTEDSLRLSWX 1 X 2 X 3 X 4 X 5 X 6 X 7 X 8 SFLIQYQESEKVG EAINLTVPGSERSYDLTGLKPGTEYTVSIYGVX 9 X 10 X 11 X 12 X 13 SX 14 X 15 LSAEFTT; wherein X 1 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or Val; X 2 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or Val; X 3 Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile, Leu, Lys, Phe, Pro, Ser, Thr,
  • TCL9 (randomized FG loop) (SEQ ID NO: 6) LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFLIQYQESEKVGEAIVLTVP GSERSYDLTGLKPGTEYTVSIYGV XX 2 X 3 X 4 X 5 X 6 X 7 X 8 X 9 X 10 X 11 X 12 SNPLSAIFTT;
  • X 1 , X 2 , X 3 , X 4 , X 5 , X 6 and X 7 is A, D, E, F, G, H, I, K, L, N, P, Q, R, S, T, V, W or Y;
  • X 8 , X 9 , X 10 , X 11 and X 12 is A, D, E, F, G, H, I, K, L, N, P, Q, R, S, T, V, W, Y or deleted.
  • DNA fragments encoding randomized BC loops (lengths 6-9 positions) or FG loops (lengths 7-12 positions) were synthesized using Slonomics technology (Sloning Biotechnology GmbH) so as to control the amino acid distribution of the library and to eliminate stop codons.
  • Slonomics technology Saoning Biotechnology GmbH
  • Two different sets of DNA molecules randomizing either the BC loop or the FG loops were synthesized independently and later combined using PCR to produce the full library product.
  • a set of synthetic DNA molecules consisting of a 5′ Tac promoter followed by the complete gene sequence of Tencon with the exception of randomized codons in the FG loop was produced (SEQ ID NOs: 26-31).
  • SEQ ID NOs: 26-31 For FG loop randomization, all amino acids except cysteine and methionine were encoded at equal percentages. The lengths of the diversified portion are such that they encode for 7, 8, 9, 10, 11, or 12 amino acids in the FG loop.
  • Sub-libraries of each length variation were synthesized individually at a scale of 2 ug and then amplified by PCR using oligos Sloning-FOR (SEQ ID NO: 9) and Sloning-Rev (SEQ ID NO: 10).
  • the 3′ fragment of the library is a constant DNA sequence containing elements for display, including a PspOMI restriction site, the coding region of the repA gene, and the cis- and ori elements.
  • PCR reactions were performed to amplify this fragment using a plasmid (pCR4Blunt) (Invitrogen) as a template with M13 Forward and M13 Reverse primers.
  • the resulting PCR products were digested by PspOMI overnight and gel-purified.
  • the TCL7 library provides for a library with randomized Tencon BC and FG loops.
  • BC loops of lengths 6-9 amino acids were mixed combinatorially with randomized FG loops of 7-12 amino acids in length.
  • Synthetic Tencon fragments BC6, BC7, BC8, and BC9 (SEQ ID NOs: 13-16, respectively) were produced to include the Tencon gene encoding for the N-terminal portion of the protein up to and including residue VX such that the BC loop is replaced with either 6, 7, 8, or 9 randomized amino acids.
  • These fragments were synthesized prior to the discovery of L17A, N46V and E831 mutations (CEN5243) but these mutations were introduced in the molecular biology steps described below. In order to combine this fragment with fragments encoding for randomized FG loops, the following steps were taken.
  • DNA fragment encoding the Tac promoter and the 5′ sequence of Tencon up to the nucleotide encoding for amino acid A17 was produced by PCR using oligos POP2222ext (SEQ ID NO: 18) and LS1114 (SEQ ID NO: 19). This was done to include the L17A mutation in the library (CEN5243).
  • DNA fragments encoding for Tencon residues R18-V75 including randomized BC loops were amplified by PCR using BC6, BC7, BC8, or BC9 as a templates and oligos LS1115 (SEQ ID NO: 20) and LS1117 (SEQ ID NO: 21).
  • This PCR step introduced a Bsal site at the 3′ end. These DNA fragments were subsequently joined by overlapping PCR using oligos POP2222ext and LS1117 as primers. The resulting PCR product of 240 bp was pooled and purified by Qiagen PCR purification kit. The purified DNA was digested with Bsal-HF and gel purified.
  • Fragments encoding the FG loop were amplified by PCR using FG7, FG8, FG9, FG10, FG11, and FG12 as templates with oligonucleotides SDG10 (SEQ ID NO: 22) and SDG24 (SEQ ID NO: 23) to incorporate a Bsal restriction site and N46V and E86I variations (CEN5243).
  • the digested BC fragments and FG fragments were ligated together in a single step using a 3-way ligation.
  • Four ligation reactions in the 16 possible combinations were set up, with each ligation reaction combining two BC loop lengths with 2 FG loop lengths.
  • Each ligation contained—300 ng of total BC fragment and 300 ng of the FG fragment.
  • These 4 ligation pools were then amplified by PCR using oligos POP2222 (SEQ ID NO: 24) and SDG28 SEQ ID N: 25). 7.5 l ag of each reaction product were then digested with Notl and cleaned up with a Qiagen PCR purification column.
  • an ankyrin repeat scaffold protein that was selected to bind to MBP was found to have a much more planar interaction surface and to bind to the outer surface of MBP distant from the active (Binz et al., Nat Biotechnol 22: 575-582, 2004).
  • shape of the binding surface of a scaffold molecule may dictate what target proteins or specific epitopes on those target proteins are able to be bound effectively by the scaffold.
  • Published efforts around engineering protein scaffolds containing FN3 domains for protein binding has relied on engineering adjacent loops for target binding, thus producing curved binding surfaces. This approach may limit the number of targets and epitopes accessible by such scaffolds.
  • Tencon and other FN3 domains contain two sets of CDR-like loops lying on the opposite faces of the molecule, the first set formed by the BC, DE, and FG loops, and the second set formed by the AB, CD, and EF loops.
  • the two sets of loops are separated by the beta-strands that form the center of the FN3 structure. If the image of the Tencon is rotated by 90 degrees, an alternative surface can be visualized. This slightly concave surface is formed by the CD and FG loops and two antiparallel beta- strands, the C and the F beta-strands, and is herein called the C-CD-F-FG surface.
  • the C-CD-F-FG surface can be used as a template to design libraries of protein scaffold interaction surfaces by randomizing a subset of residues that form the surface.
  • Beta-strands have a repeating structure with the side chain of every other residue exposed to the surface of the protein.
  • a library can be made by randomizing some or all surface exposed residues in the beta strands.
  • TCL14 library (SEQ ID NO: 7): LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFX 1 IX 2 YX 3 EX 4 X 5 X 6 X 7 GE AIVLTVPGSERSYDLTGLKPGTEYX 8 VX 9 IX 10 GVKGGX 11 X 12 SX 13 PL SAIFTT; wherein X 1 , X 2 , X 3 , X 4 , X 5 , X 6 , X 7 , X 8 , X 9 , X 10 , X 11 , X 12 and X 13 are A, D, E, F, G, H, I, K, L, N, P, Q, R, S, T, V, W, Y, C or M.
  • the two beta strands forming the C-CD-F-FG surface in Tencon27 have a total of 9 surface exposed residues that could be randomized; C-strand: S30, L32, Q34, Q36; F-strand: E66, T68, S70, Y72, and V74, while the CD loop has 6 potential residues: S38, E39, K40, V41, G42, and E43 and the FG loop has 7 potential residues: K75, G76, G77, H78, R79, S80, and N81. Select residues were chosen for inclusion in the TCL14 design due to the larger theoretical size of the library if all 22 residues were randomized.
  • TCL19, TCL21 and TCL23 are randomized at the same positions as TCL14 (see above) however the distribution of amino acids occurring at these positions is altered (Table 3).
  • TCL19 and TCL21 were designed to include an equal distribution of 18 natural amino acids at every position (5.55% of each), excluding only cysteine and methionine.
  • TCL23 was designed such that each randomized position approximates the amino acid distribution found in the HCDR3 loops of functional antibodies (Birtalan et al., J Mol Biol 377: 1518-1528, 2008) as described in Table 3.
  • cysteine and methionine were excluded.
  • TCL24 4 additional Tencon positions were randomized as compared to libraries TCL14, TCL19, TCL21, and TCL23. These positions include N46 and T48 from the D strand and S84 and 186 from the G strand. Positions 46, 48, 84, and 86 were chosen in particular as the side chains of these residues are surface exposed from beta-strands D and G and lie structurally adjacent to the randomized portions of the C and F strand, thus increasing the surface area accessible for binding to target proteins.
  • the amino acid distribution used at each position for TCL24 is identical to that described for TCL19 and TCL21 in Table 3.
  • TCL24 Library (SEQ ID NO: 8) LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFX 1 IX 2 YX 3 EX 4 X 5 X 6 X 7 GE AIX 8 LX 9 VPGSERSYDLTGLKPGTEYX 10 VX 11 IX 12 GVKGGX 13 X 14 S X 15 PLX 16 AX 17 FTT; wherein X 1 , X 2 , X 3 , X 4 , X 5 , X 6 , X 10 , X 11 , X 12 , X 13 , X 14 , X 15 , X 16 and X 17 are A, D, E, F, G, H, I, K, L, N, P, Q, R, S, T, V, Y or W.
  • the TCL21 library was generated using Colibra library technology (Isogenica) in order to control amino acid distributions.
  • TCL19, TCL23, and TCL24 gene fragments were generated using Slonomics technology (Morphosys) to control amino acid distributions.
  • PCR was used to amplify each library following initial synthesis followed by ligation to the gene for RepA in order to be used in selections using the CIS-display system (Odegrip et al., Proc Natl Acad Sci U S A 101: 2806-2810, 2004) as described above for the loop libraries.
  • FN3 domains specific for human PD-L1 were selected via CIS-Display (Odegrip et al 2004) using recombinant biotinylated PD-L1 (rhPD-L1/Fc Chimera, R&D Systems 156-B7).
  • ITT in vitro transcription and translation
  • 3 ⁇ g of DNA from libraries TCL18, TCL19, TCL21, TCL23, and TCL24 were incubated at 30° C. with 0.1 mM complete amino acids, 1X S30 premix components, and 15 ⁇ L of S30 extract (Isogenica) in a total volume of 50 ⁇ L.
  • Outputs from the fifth round panning were subjected to four additional rounds of off-rate selection.
  • Library transcription and translation was performed as described above after which the ITT reactions were incubated with biotinylated recombinant PD-L1 proteins and captured on neutravidin or streptavidin coated magnetic beads, before being washed in TBST extensively then subsequently washed in 5 ⁇ M cold recombinant PD-L1 protein for 1 hour.
  • the biotinylated target antigen concentration was reduced from 25 nM in rounds 6 and 7 to 2.5 nM in rounds 8 and 9.
  • genes encoding the selected FN3 domains were amplified by PCR, subcloned into a pET vector modified to include a ligase independent cloning site, and transformed into BL21 (DE3) (Stratagene) cells for soluble expression in E. coli using standard molecular biology techniques.
  • a gene sequence encoding a C-terminal poly-histidine tag was added to each FN3 domain to enable purification and detection.
  • Cultures were grown to an optical density of 0.6-0.8 in TB medium supplemented with 100 ⁇ g/mL carbenicillin in 1 mL 96-well blocks at 37° C. before the addition of IPTG to 1 mM, at which point the temperature was reduced to 30° C.
  • Cells were harvested approximately 16 hours later by centrifugation and frozen at ⁇ 20° C. Cell lysis was achieved by incubating each pellet in 0.6 mL of BugBuster® HT lysis buffer (Novagen EMD Biosciences) supplemented with 0.2 mg/mL lysozyme with shaking at room temperature for 30 minutes.
  • BugBuster® HT lysis buffer Novagen EMD Biosciences
  • Streptavidin-coated Maxisorp plates (Nunc catalog 436110) were blocked for lh in Starting Block T20 (Pierce) and then coated with biotinylated PD-L1 (using same antigen as in panning) or negative controls (an unrelated Fc-fused recombinant protein and human serum albumin) for lh. Plates were rinsed with TBST and diluted lysate was applied to plates for 1h. Following additional rinses, wells were treated with HRP-conjugated anti-FN3 domain antibody (PAB25) for lh and then assayed with POD (Roche catalog 11582950001).
  • POD HRP-conjugated anti-FN3 domain antibody
  • Size exclusion chromatography was used to determine the aggregation state of anti-PD-L1 FN3 domains Aliquots (10)(L) of each purified FN3 domain were injected onto a Superdex 75 5/150 column (GE Healthcare) at a flow rate of 0.3 mL/min in a mobile phase of PBS pH 7.4. Elution from the column was monitored by absorbance at 280 nm. Tencon protein was included in each run as a control. Agilent ChemStation software was used to analyse the elution profiles. 20 anti-PD-L1 FN3 domains demonstrated a retention time between 5.2 and 6.4 minutes and only a single SEC peak indicative of monomeric protein (Table 6).
  • SEQ ID No. 1 Original Tencon Sequence LPAPKNLVVSEVTEDSLRLSWTAPDAAFDSFLIQYQESEKVGEAINLTVPGSERSY DLTGLKPGTEYTVSIYGVKGGHRSNPLSAEFTT SEQ ID No.
  • X 1 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or Val
  • X 2 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or Val
  • X 3 Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or Val
  • SEQ ID No. 4 Stabilized Tencon LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFLIQYQESEKVGEAIVLTVPGSERSY DLTGLKPGTEYTVSIYGVKGGHRSNPLSAIFTT SEQ ID No.
  • TCL14 library (SEQ ID NO: 7): LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFX 1 IX 2 YX 3 EX 4 X 5 X 6 X 7 GEAIVLTVPGS ERSYDLTGLKPGTEYX 8 VX 9 IX 10 GVKGGX 11 X 12 SX 13 PLSAIFTT; wherein X 1 , X 2 , X 3 , X 4 , X 5 , X 6 , X 7 , X 8 , X 9 , X 10 , X 11 , X 12 and X 13 are A, D, E, F, G, H, I, K, L, N, P, Q, R, S, T, V, W, Y, C or M.
  • TCL24 Library (SEQ ID NO: 8) LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFX 1 IX 2 YX 3 EX 4 X 5 X 6 X 7 GEAIX 8 LX 9 VPG SERSYDLTGLKPGTEYX 10 VX 11 IX 12 GVKGGX 13 X 14 SX 15 PLX 16 AX 17 FTT; wherein X 1 , X 2 , X 3 , X 4 , X 5 , X 6 , X 10 , X 11 , X 12 , X 13 , X 14 , X 15 , X 16 and X 17 are A, D, E, F, G, H, I, K, L, N, P, Q, R, S, T, V, Y or W.
  • SEQ ID No. 9 Sloning-FOR GTGACACGGCGGTTAGAAC
  • SEQ ID No. 10 Sloning-REV GCCTTTGGGAAGCTTCTAAG
  • SEQ ID No. 11 POP2250 CGGCGGTTAGAACGCGGCTACAATTAATAC
  • SEQ ID No. 12 DigLigRev CATGATTACGCCAAGCTCAGAA SEQ ID No.

Abstract

FN3 domains that specifically bind to PD-L1, their conjugates, isolated nucleotides encoding the molecules, vectors, host cells, and methods of making and using them are useful in therapeutic and diagnostic applications.

Description

CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Application Ser. No. 62/434,054, filed 14 Dec. 2016, the entire contents of which are incorporated herein by reference.
SEQUENCE LISTING
This application contains a Sequence Listing submitted via EFS-Web, the entire content of which is incorporated herein by reference. The ASCII text file, created on 11 Dec. 2017, is named JBI5113USNP_ST25.txt and is 124 kilobytes in size.
FIELD OF THE INVENTION
The present invention relates to fibronectin type III domains that specifically bind to PD-L1 and methods of making and using the molecules.
BACKGROUND OF THE INVENTION
The immune system is tightly controlled by a network of costimulatory and co-inhibitory ligands and receptors. These molecules provide secondary signals for T cell activation and provide a balanced network of positive and negative signals to maximize immune responses against infection and tumors, while limiting immunity to self (Wang et al., (Epub Mar. 7, 2011) J Exp Med 208(3):577-92; Lepenies et al., (2008) Endocr Metab Immune Disord Drug Targets 8:279-288).
Programmed Death-1 (PD-1) is a key immune checkpoint receptor expressed by activated T and B cells and mediates immunosuppression. The ligand for PD-1, PD-L1, is expressed by antigen-presenting cells and many cancers such as lung, ovarian and colon carcinoma and various myelomas. Binding of PD-L1 to PD-1 on T cells downregulates T cell proliferation and activation and drives T cell anergy and exhaustion in the tumor microenvironment, facilitating tumor cell escape from T-cell mediated immune surveillance.
Therapeutic efficacy of PD-1 and PD-L1 antagonists has been validated in clinical trials. However, response rates remain low. For example, Opdivo® (Nivolumab) treatment achieved a 26% objective response rate (ORR) across the 27 clinical trials analyzed (Tie et al., Int J Cancer 2016 Nov 4 doi: 10.1002/ijc.30501. [Epub ahead of print])
Measuring the expression of PD-L1 protein in the tumor tissue may aid in the early detection of cancer pathologies and may help assess the efficacy and durability of PD-L1 and PD-1 antagonists. For example, PD-L1 expression in at least 50% of tumor cells correlated with improved efficacy of Keytruda® (pembrolizumab) (Garon et al., N Engl J Med 2015; 372:2018-2028), and PD-L1 expression has been correlated with poor prognosis (see for example Wang et al., Eur J Surg oncol 2015 Apr; 41(4):450-6).
However, the use of PD-L1 protein expression as an accurate predictor for cancer and/or the efficacy of anti-PD-1 and anti-PD-L1 directed therapies remain challenging partially due to observed variability in results depending on the detection reagent used. For example, the evaluation of PD-L1 expression in non-small cell lung cancer samples using commercially available assays such as PD-L1 (E1L3NO) XP® Rabbit mAb (Cell Signaling) and Ventana PD-L1 (SP142) Assay yielded discordant results (McLaughlin et al., JAMA Oncol 2016 Jan;2(1):46-54)
Therefore, there is a need for reagents to accurately detect PD-L1 in tumor tissues and other samples and new therapeutics that modulate the interaction between PD-L1 and PD-1.
SUMMARY OF THE INVENTION
The invention provides an isolated FN3 domain that specifically binds to PD-L1.
The invention also provides an isolated FN3 domain that specifically binds to PD-L1 comprising the sequence of SEQ ID NOs: 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123 or 124.
The invention also provides an isolated polynucleotide encoding the FN3 domain that specifically binds to PD-L1 of the invention.
The invention also provides a vector comprising the polynucleotide of the invention.
The invention also provides a host cell comprising the vector of the invention.
The invention also provides a method of producing the FN3 domain that specifically binds to PD-L1 of the invention, comprising culturing the isolated host cell of the invention under conditions that the FN3 domain that specifically binds to PD-L1 is expressed, and purifying the FN3 domain that specifically binds to PD-L1.
The invention also provides a pharmaceutical composition comprising the FN3 domain that specifically binds to PD-L1 of the invention and a pharmaceutically acceptable carrier.
The invention also provides an anti-idiotypic antibody that specifically binds the FN3 domain that specifically binds to PD-L1 of the invention.
The invention also provides a kit comprising the FN3 domain of the invention.
The invention also provides a method of detecting PD-L1-expressing cancer cells in a tumor tissue, comprising
    • obtaining a sample of the tumor tissue from a subject; and
    • detecting whether PD-L1 is expressed in the tumor tissue by contacting the sample of the tumor tissue with the FN3 domain that specifically binds to PD-L1 comprising the sequence of any one of SEQ ID NOs: 34-124 and detecting the binding between PD-L1 and the FN3 domain.
The invention also provides a method of isolating or detecting PD-L1 expressing cells, comprising obtaining a sample from a subject;
    • contacting the sample with the FN3 domain that specifically binds to PD-L1
    • comprising the sequence of any one of SEQ ID NOs: 34-124, and
    • isolating or detecting the cells bound to the FN3 domains.
The invention also provides a method of detecting PD-L1-expressing cancer cells in a tumor tissue, comprising
    • conjugating the FN3 domain that specifically binds to PD-L1 comprising the sequence of any one of SEQ ID NOs: 34-124 to a detectable label to form a conjugate;
    • administering the conjugate to a subject; and
    • visualizing the PD-L1 expressing cancer cells to which the conjugate is bound.
DETAILED DESCRIPTION OF THE INVENTION
As used in this specification and the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the content clearly dictates otherwise. Thus, for example, reference to “a cell” includes a combination of two or more cells, and the like.
“Fibronectin type III (FN3) domain” (FN3 domain) refers to a domain occurring frequently in proteins including fibronectins, tenascin, intracellular cytoskeletal proteins, cytokine receptors and prokaryotic enzymes (Bork and Doolittle, Proc Nat Acad Sci USA 89:8990-8994, 1992; Meinke et al., J Bacteriol 175:1910-1918, 1993; Watanabe et al., J Biol Chem 265:15659-15665, 1990). Exemplary FN3 domains are the 15 different FN3 domains present in human tenascin C, the 15 different FN3 domains present in human fibronectin (FN), and non-natural synthetic FN3 domains as described for example in U.S. Pat. No. 8,278,419. Individual FN3 domains are referred to by domain number and protein name, e.g., the 3rd FN3 domain of tenascin (TN3), or the 10th FN3 domain of fibronectin (FN10).
“Centyrin” refers to a FN3 domain that is based on the consensus sequence of the 15 different FN3 domains present in human tenascin C.
The term “capture agent” refers to substances that bind to a particular type of cells and enable the isolation of that cell from other cells. Exemplary capture agents are magnetic beads, ferrofluids, encapsulating reagents, molecules that bind the particular cell type and the like.
“Sample” refers to a collection of similar fluids, cells, or tissues isolated from a subject, as well as fluids, cells, or tissues present within a subject. Exemplary samples are tissue biopsies, fine needle aspirations, surgically resected tissue, organ cultures, cell cultures and biological fluids such as blood, serum and serosal fluids, plasma, lymph, urine, saliva, cystic fluid, tear drops, feces, sputum, mucosal secretions of the secretory tissues and organs, vaginal secretions, ascites fluids, fluids of the pleural, pericardial, peritoneal, abdominal and other body cavities, fluids collected by bronchial lavage, synovial fluid, liquid solutions contacted with a subject or biological source, for example, cell and organ culture medium including cell or organ conditioned medium and lavage fluids and the like.
“Substituting” or “substituted” or “mutating” or “mutated” refers to altering, deleting of inserting one or more amino acids or nucleotides in a polypeptide or polynucleotide sequence to generate a variant of that sequence.
“Variant” refers to a polypeptide or a polynucleotide that differs from a reference polypeptide or a reference polynucleotide by one or more modifications for example, substitutions, insertions or deletions.
“Specifically binds” or “specific binding” refers to the ability of the FN3 domain of the invention to bind PD-L1 with a dissociation constant (KD) of about 1×10−6 M or less, for example about 1×10−7 M or less, about 1×10−8 M or less, about 1×10−9 M or less, about 1×10−10 M or less, about 1×10−11 M or less, about 1×10−12 M or less, or about 1×10−13 M or less. Alternatively, “specific binding” refers to the ability of the FN3 domain of the invention to bind PD-L1 at least 5-fold above the negative control in standard ELISA assay. The isolated FN3 domain of the invention that specifically binds PD-L1 may, however, have cross-reactivity to other related antigens, for example to the same predetermined antigen from other species (homologs), such as Macaca Fascicularis (cynomolgous monkey, cyno) or Pan troglodytes (chimpanzee).
“Library” refers to a collection of variants. The library may be composed of polypeptide or polynucleotide variants.
“Stability” refers to the ability of a molecule to maintain a folded state under physiological conditions such that it retains at least one of its normal functional activities, for example, binding to a predetermined antigen such as PD-L1.
“PD-L1” refers to human PD-L1 protein having the amino acid sequence of SEQ ID NO: 32. The extracellular domain of PD-L1 spans residues 1-220, the transmembrane domain spans residues 221-241 and the cytoplasmic domain spans residues 242-272.
“PD-1” refers to human PD-1 protein having the amino acid sequence of SEQ ID NO: 33. The extracellular domain of PD-1 spans residues 1-150, the transmembrane domain spans residues 151-171 and the cytoplasmic domain spans residues 172-268 of SEQ ID NO: 33.
“Tencon” refers to the synthetic fibronectin type III (FN3) domain having the sequence shown in SEQ ID NO: 1 and described in U.S. Pat. Publ. No. 2010/0216708.
A “cancer cell” or a “tumor cell” refers to a cancerous, pre-cancerous or transformed cell, either in vivo, ex vivo, and in tissue culture, that has spontaneous or induced phenotypic changes that do not necessarily involve the uptake of new genetic material. Although transformation can arise from infection with a transforming virus and incorporation of new genomic nucleic acid, or uptake of exogenous nucleic acid, it can also arise spontaneously or following exposure to a carcinogen, thereby mutating an endogenous gene. Transformation/cancer is exemplified by, e.g., morphological changes, immortalization of cells, aberrant growth control, foci formation, proliferation, malignancy, tumor specific markers levels, invasiveness, tumor growth or suppression in suitable animal hosts such as nude mice, and the like, in vitro, in vivo, and ex vivo (Freshney, Culture of Animal Cells: A Manual of Basic Technique (3rd ed. 1994)).
“Vector” refers to a polynucleotide capable of being duplicated within a biological system or that can be moved between such systems. Vector polynucleotides typically contain elements, such as origins of replication, polyadenylation signal or selection markers that function to facilitate the duplication or maintenance of these polynucleotides in a biological system. Examples of such biological systems may include a cell, virus, animal, plant, and reconstituted biological systems utilizing biological components capable of duplicating a vector. The polynucleotide comprising a vector may be DNA or RNA molecules or a hybrid of these.
“Expression vector” refers to a vector that can be utilized in a biological system or in a reconstituted biological system to direct the translation of a polypeptide encoded by a polynucleotide sequence present in the expression vector.
“Polynucleotide” refers to a synthetic molecule comprising a chain of nucleotides covalently linked by a sugar-phosphate backbone or other equivalent covalent chemistry. cDNA is a typical example of a polynucleotide.
“Polypeptide” or “protein” refers to a molecule that comprises at least two amino acid residues linked by a peptide bond to form a polypeptide. Small polypeptides of less than about 50 amino acids may be referred to as “peptides”.
“Valent” refers to the presence of a specified number of binding sites specific for an antigen in a molecule. As such, the terms “monovalent”, “bivalent”, “tetravalent”, and “hexavalent” refer to the presence of one, two, four and six binding sites, respectively, specific for an antigen in a molecule.
“Subject” includes any human or nonhuman animal “Nonhuman animal” includes all vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dogs, cats, horses, cows chickens, amphibians, reptiles, etc. Except when noted, the terms “patient” or “subject” are used interchangeably.
“Isolated” refers to a homogenous population of molecules (such as synthetic polynucleotides or a polypeptide such as FN3 domains) which have been substantially separated and/or purified away from other components of the system the molecules are produced in, such as a recombinant cell, as well as a protein that has been subjected to at least one purification or isolation step. “Isolated FN3 domain” refers to an FN3 domain that is substantially free of other cellular material and/or chemicals and encompasses FN3 domains that are isolated to a higher purity, such as to 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% purity.
Compositions of Matter
The present invention provides fibronectin type III (FN3) domains that specifically bind PD-L1. These molecules can be used in therapeutic and diagnostic applications and in imaging. The present invention provides polynucleotides encoding the FN3 domains of the invention or complementary nucleic acids thereof, vectors, host cells, and methods of making and using them.
The invention provides an isolated FN3 domain that specifically binds PD-L1.
The FN3 domain of the invention may bind PD-L1 with a dissociation constant (KD) of less than about 1×10−7 M, for example less than about 1×10−8 M, less than about 1×10−9 M, less than about 1×10−1° M, less than about 1×10−11 M, less than about 1×10−12 M, or less than about 1×10−13 M as determined by surface plasmon resonance or the Kinexa method, as practiced by those of skill in the art. The measured affinity of a particular FN3 domain-antigen interaction can vary if measured under different conditions (e.g., osmolarity, pH). Thus, measurements of affinity and other antigen-binding parameters (e.g., KD, Kon, Koff) are made with standardized solutions of protein scaffold and antigen, and a standardized buffer, such as the buffer described herein.
The FN3 domain of the invention may bind PD-L1 at least 5-fold above the signal obtained for a negative control in standard ELISA assay.
In some embodiments, the FN3 domain that specifically binds PD-L1 comprises an initiator methionine (Met) linked to the N-terminus of the molecule.
In some embodiments, the FN3 domain that specifically binds PD-L1 comprises a cysteine (Cys) linked to a C-terminus of the FN3 domain.
The addition of the N-terminal Met and/or the C-terminal Cys may facilitate expression and/or conjugation of half-life extending molecules.
In some embodiments, the FN3 domain that specifically binds PD-L1 is internalized into a cell.
Internalization of the FN3 domain may facilitate delivery of a cytotoxic agent into tumor cells.
In some embodiments, the FN3 domain that specifically binds PD-L1 inhibits binding of PD-L1 to PD-1.
Inhibition of binding of PD-L1 to PD-1 by the FN3 domains of the invention may be assessed using competition ELISA. In an exemplary assay, 1 μg/ml recombinant human PD-L1 extracellular domain is bound on wells of microtiter plates, the wells are washed and blocked, and 10 μg/ml of the test FN3 domain is added. Without washing, 7.5 μg/ml PD-1 extracellular domain is added into the wells and incubated for 30 min, after which 0.5 μg/ml anti-PD-1 antibody is added and incubated for 30 min. The plates are washed and 0.5 μg/mL neutravidin-HRP conjugate polyclonal antibody is added and incubated for 30 minutes. The plates are washed and POD Chemiluminescence substrate added immediately prior to reading the luminescence signal. The FN3 domains of the invention inhibit binding of PD-L1 to PD-1 when the binding of PD-1 is reduced by at least about 80%, 85%, 90%, 95% or 100%.
In some embodiments, the FN3 domain that specifically binds PD-L1 is a PD-L1 antagonist.
In some embodiments, the FN3 domain that specifically binds PD-L1 is a PD-L1 agonist.
“Antagonist” refers to a FN3 domain that specifically binds PD-L1 that suppresses at least one reaction or activity that is induced by PD-L1 binding PD-1. A molecule is an antagonist when the at least one reaction or activity is suppressed by at least about 30%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% more than the at least one reaction or activity suppressed in the absence of the antagonist (e.g., negative control), or when the suppression is statistically significant when compared to the suppression in the absence of the antagonist. A typical reaction or activity that is induced by PD-L1 binding PD-1 is reduced antigen-specific CD4+ or CD8+ cell proliferation or reduced interferon-γ (IFN-γ) production by T cells.
The antagonistic FN3 domains that specifically bind PD-L1 may be used in the treatment of cancer or viral infections and in general in treatment of diseases in which activation of immune responses is desirable.
“Agonist” refers to a FN3 domain that specifically binds PD-L1 that induces at least one reaction or activity that is induced by PD-L1 binding PD-1. The FN3 domain is an agonist when the at least one reaction or activity is induced by at least about 30%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% greater than the at least one reaction or activity induced in the absence of the agonist (e.g., negative control), or when the induction is statistically significant when compared to the induction in the absence of the agonist. A typical reaction or activity that is induced by PD-L1 binding PD-1 is reduced antigen-specific CD4+ or CD8+ cell proliferation or reduced interferon-γ (IFN-γ) production by T cells.
The agonistic FN3 domains that specifically bind PD-L1 may be used in the treatment of autoimmune or inflammatory diseases and in general diseases in which suppression of immune responses is desirable.
In some embodiments, the FN3 domain that specifically binds PD-L1 does not inhibit binding of PD-L1 to PD-1.
In some embodiments, the FN3 domain that specifically binds PD-L1 does not activate signaling downstream of PD-1.
In some embodiments, the FN3 domain that specifically binds PD-L1 is based on Tencon sequence of SEQ ID NO: 1 or Tencon 27 sequence of SEQ ID NO: 4, optionally having substitutions at residues positions 11, 14, 17, 37, 46, 73, and/or 86 (residue numbering corresponding to SEQ ID NO: 4).
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NOs: 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123 and/or 124.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 34.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 35.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 36.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 37.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 38.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 39.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 40.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 41.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 42.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 43.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 44.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 45.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 46.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 47.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 48.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 49.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 50.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 51.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 52.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 53.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 54.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 55.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 56.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 57.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 58.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 59.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 60.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 61.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 62.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 63.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 64.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 65.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 66.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 67.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 68.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 69.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 70.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 71.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 72.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 73.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 74.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 75.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 76.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 77.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 78.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 79.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 80.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 81.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 82.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 83.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 84.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 85.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 86.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 87.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 88.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 89.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 90.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 91.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 92.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 93.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 94.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 95.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 96.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 97.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 98.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 99.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 100.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 101.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 102.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 103.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 104.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 105.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 106.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 107.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 108.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 109.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 110.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 111.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 112.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 113.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 114.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 115.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 116.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 117.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 118.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 119.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 120.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 121.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 122.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 123.
The invention also provides an isolated FN3 domain that specifically binds PD-L1 comprising the amino acid sequence of SEQ ID NO: 124.
In some embodiments, the isolated FN3 domain that specifically binds PD-L1 comprises an initiator methionine (Met) linked to the N-terminus of the molecule.
in some embodiments, the isolated FN3 domain that specifically binds PD-L1 comprises an amino acid sequence that is 62%, 63%, 64% , 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO: 74.
In some embodiments, the isolated FN3 domain that specifically binds PD-L1 comprises an amino acid sequence that is 62%, 63%, 64% , 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of any one of SEQ ID NOs: 34-124.
Conjugates of the FN3 Domains that Specifically Bind PD-L1 of the Invention
The invention also provides an isolated FN3 domain that specifically binds PD-L1 conjugated to a heterologous molecule(s).
In some embodiments, the heterologous molecule is a detectable label or a cytotoxic agent.
The invention also provides an FN3 domain that specifically binds PD-L1 conjugated to a detectable label.
The invention also provides an FN3 domain that specifically binds PD-L1 conjugated to a cytotoxic agent.
In some embodiments, the detectable label is also a cytotoxic agent.
The FN3 domains that specifically bind PD-L1 of the invention conjugated to a detectable label can be used to evaluate expression of PD-L1 on samples such as tumor tissue in vivo or in vitro.
Detectable label includes compositions that when conjugated to the FN3 domains that specifically bind PD-L 1 of the invention renders the latter detectable, via spectroscopic, photochemical, biochemical, immunochemical, or chemical means.
Exemplary detectable labels include radioactive isotopes, magnetic beads, metallic beads, colloidal particles, fluorescent dyes, electron-dense reagents, enzymes (for example, as commonly used in an ELISA), biotin, digoxigenin, haptens, luminescent molecules, chemiluminescent molecules, fluorochromes, fluorophores, fluorescent quenching agents, colored molecules, radioactive isotopes, scintillants, avidin, streptavidin, protein A, protein G, antibodies or fragments thereof, poly histidine, Ni2+, Flag tags, myc tags, heavy metals, enzymes, alkaline phosphatase, peroxidase, luciferase, electron donors/acceptors, acridinium esters, and colorimetric substrates.
A detectable label may emit a signal spontaneously, such as when the detectable label is a radioactive isotope. In other cases the detectable label emits a signal as a result of being stimulated by an external field.
Exemplary radioactive isotopes may be y-emitting, Auger-emitting, β-emitting, an alpha-emitting or positron-emitting radioactive isotope. Exemplary radioactive isotopes include 3H, 11C, 13C, 15N, 18F, 19F, 55Co, 57Co, 60Co, 61Cu, 62Cu, 64Cu, 67Cu, 68Cu, 72As, 75Br, 86Y, 89Zr, 90Sr, 94mTc, 99mTc, 115In, 123I, 124I, 125I, 131I, 211At, 212Bi, 213Bi, 223Ra, 226Ra, 225Ac and 227Ac.
Exemplary metal atoms are metals with an atomic number greater than 20, such as calcium atoms, scandium atoms, titanium atoms, vanadium atoms, chromium atoms, manganese atoms, iron atoms, cobalt atoms, nickel atoms, copper atoms, zinc atoms, gallium atoms, germanium atoms, arsenic atoms, selenium atoms, bromine atoms, krypton atoms, rubidium atoms, strontium atoms, yttrium atoms, zirconium atoms, niobium atoms, molybdenum atoms, technetium atoms, ruthenium atoms, rhodium atoms, palladium atoms, silver atoms, cadmium atoms, indium atoms, tin atoms, antimony atoms, tellurium atoms, iodine atoms, xenon atoms, cesium atoms, barium atoms, lanthanum atoms, hafnium atoms, tantalum atoms, tungsten atoms, rhenium atoms, osmium atoms, iridium atoms, platinum atoms, gold atoms, mercury atoms, thallium atoms, lead atoms, bismuth atoms, francium atoms, radium atoms, actinium atoms, cerium atoms, praseodymium atoms, neodymium atoms, promethium atoms, samarium atoms, europium atoms, gadolinium atoms, terbium atoms, dysprosium atoms, holmium atoms, erbium atoms, thulium atoms, ytterbium atoms, lutetium atoms, thorium atoms, protactinium atoms, uranium atoms, neptunium atoms, plutonium atoms, americium atoms, curium atoms, berkelium atoms, californium atoms, einsteinium atoms, fermium atoms, mendelevium atoms, nobelium atoms, or lawrencium atoms.
In some embodiments, the metal atoms may be alkaline earth metals with an atomic number greater than twenty.
In some embodiments, the metal atoms may be lanthanides.
In some embodiments, the metal atoms may be actinides.
In some embodiments, the metal atoms may be transition metals.
In some embodiments, the metal atoms may be poor metals.
In some embodiments, the metal atoms may be gold atoms, bismuth atoms, tantalum atoms, and gadolinium atoms.
In some embodiments, the metal atoms may be metals with an atomic number of 53 (i.e. iodine) to 83 (i.e. bismuth).
In some embodiments, the metal atoms may be atoms suitable for magnetic resonance imaging.
The metal atoms may be metal ions in the form of +1 , +2, or +3 oxidation states, such as Ba2+, Bi3+, Cs+, Ca2+, Cr2+, Cr3+, Cr6+, Co3+, Co3+, Cu+, Cu2+, Cu3+, Ga3+, Gd3+, Au+, Au3+, Pe2+, Fe3+, F3+, Pb2+, Mn2+, Mn3+, Mn4+, Mn7+, Hg2+, Ni2+, Ni3+, Ag+, Sr2+, Sn2+, Sn4+, and Zn2+. The metal atoms may comprise a metal oxide, such as iron oxide, manganese oxide, or gadolinium oxide.
Suitable dyes include any commercially available dyes such as, for example, 5(6)-carboxyfluorescein, IRDye 680RD maleimide or IRDye 800CW, ruthenium polypyridyl dyes, and the like.
Suitable fluorophores are fluorescein isothiocyante (FITC), fluorescein thiosemicarbazide, rhodamine, Texas Red, CyDyes (e.g., Cy3, Cy5, Cy5.5), Alexa Fluors (e.g., Alexa488, Alexa555, Alexa594; Alexa647), near infrared (NIR) (700-900 nm) fluorescent dyes, and carbocyanine and aminostyryl dyes.
The FN3 domains that specifically bind PD-L1 conjugated to a detectable label may be used as an imaging agent to evaluate tumor distribution, diagnosis for the presence of tumor cells and/or, recurrence of tumor.
In some embodiments, the FN3 domains that specifically bind PD-L1 of the invention are conjugated to a cytotoxic agent.
In some embodiments, the cytotoxic agent is a chemotherapeutic agent, a drug, a growth inhibitory agent, a toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
The FN3 domains that specifically bind PD-L1 conjugated to a cytotoxic agent of the invention may be used in the targeted delivery of the cytotoxic agent to PD-L1 expressing tumor cell, and intracellular accumulation therein, wherein systemic administration of these unconjugated cytotoxic agents may result in unacceptable levels of toxicity to normal cells.
In some embodiments, the cytotoxic agent is daunomycin, doxorubicin, methotrexate, vindesine, bacterial toxins such as diphtheria toxin, ricin, geldanamycin, maytansinoids or calicheamicin. The cytotoxic agent may elict their cytotoxic and cytostatic effects by mechanisms including tubulin binding, DNA binding, or topoisomerase inhibition.
In some embodiments, the cytotoxic agent is an enzymatically active toxins such as diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes.
In some embodiments, the cytotoxic agent is a radionuclide, such as 212Bi, 131I, 131In, 90Y, and 186Re.
In some embodiments, the cytotoxic agent is dolastatins or dolostatin peptidic analogs and derivatives, auristatin or monomethyl auristatin phenylalanine Exemplary molecules are disclosed in U.S. Pat Nos. 5,635,483 and 5,780,588. Dolastatins and auristatins have been shown to interfere with microtubule dynamics, GTP hydrolysis, and nuclear and cellular division (Woyke et al (2001) Antimicrob Agents and Chemother. 45(12):3580-3584) and have anticancerand antifungal activity. The dolastatin or auristatin drug moiety may be attached to the FN3 domain of the invention through the N (amino) terminus or the C (carboxyl) terminus of the peptidic drug moiety (WO 02/088172), or via any cysteine engineered into the FN3 domain.
The FN3 domains that specifically bind PD-L1 of the invention may be conjugated to a detectable label using known methods.
In some embodiments, the detectable label is complexed with a chelating agent.
In some embodiments, the detectable label is conjugated to the FN3 domain that specifically binds PD-L1 of the invention via a linker.
The detectable label or the cytotoxic moiety may be linked directly, or indirectly, to the FN3 domain that specifically binds PD-L1 of the invention using known methods. Suitable linkers are known in the art and include, for example, prosthetic groups, non-phenolic linkers (derivatives of N-succimidyl-benzoates; dodecaborate), chelating moieties of both macrocyclics and acyclic chelators, such as derivatives of 1,4,7,10-tetraazacyclododecane-1,4,7,10,tetraacetic acid (DOTA), derivatives of diethylenetriaminepentaacetic avid (DTPA), derivatives of S-2-(4-Isothiocyanatobenzyl)-1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA) and derivatives of 1,4,8,11-tetraazacy clodocedan-1,4,8,11-tetraacetic acid (TETA), N-succinimidyl-3 -(2-pyridyldithiol) propionate (SPDP), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HC1), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as bis(p-azidobenzoyl)hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as toluene 2,6-diisocyanate), and bis-active fluorine compounds (such as 1,5-difluoro-2,4-dinitrobenzene) and other chelating moieties. Suitable peptide linkers are well known.
In some embodiment, the FN3 domain that specifically binds PD-L1 is removed from the blood via renal clearance.
Isolation of PD-L1 binding FN3 domains from a library based on Tencon sequence
Tencon (SEQ ID NO: 1) is a non-naturally occurring fibronectin type III (FN3) domain designed from a consensus sequence of fifteen FN3 domains from human tenascin-C (Jacobs et al., Protein Engineering, Design, and Selection, 25:107-117, 2012; U.S. Pat. Publ. No. 2010/0216708). The crystal structure of Tencon shows six surface-exposed loops that connect seven beta-strands as is characteristic to the FN3 domains, the beta-strands referred to as A, B, C, D, E, F, and G, and the loops referred to as AB, BC, CD, DE, EF, and FG loops (Bork and Doolittle, Proc Natl Acad Sci USA 89:8990-8992, 1992; U.S. Pat. No. 6,673,901). These loops, or selected residues within each loop, may be randomized in order to construct libraries of fibronectin type III (FN3) domains that may be used to select novel molecules that bind Pd-L1. Table 1 shows positions and sequences of each loop and beta-strand in Tencon (SEQ ID NO: 1).
Library designed based on Tencon sequence may thus have randomized FG loop, or randomized BC and FG loops, such as libraries TCL1 or TCL2 as described below. The Tencon BC loop is 7 amino acids long, thus 1, 2, 3, 4, 5, 6 or 7 amino acids may be randomized in the library diversified at the BC loop and designed based on Tencon sequence. The Tencon FG loop is 7 amino acids long, thus 1, 2, 3, 4, 5, 6 or 7 amino acids may be randomized in the library diversified at the FG loop and designed based on Tencon sequence. Further diversity at loops in the Tencon libraries may be achieved by insertion and/or deletions of residues at loops. For example, the FG and/or BC loops may be extended by 1-22 amino acids, or decreased by 1-3 amino acids. The FG loop in Tencon is 7 amino acids long, whereas the corresponding loop in antibody heavy chains ranges from 4-28 residues. To provide maximum diversity, the FG loop may be diversified in sequence as well as in length to correspond to the antibody CDR3 length range of 4-28 residues. For example, the FG loop can further be diversified in length by extending the loop by additional 1, 2, 3, 4 or 5 amino acids.
Library designed based on Tencon sequence may also have randomized alternative surfaces that form on a side of the FN3 domain and comprise two or more beta strands, and at least one loop. One such alternative surface is formed by amino acids in the C and the F beta-strands and the CD and the FG loops (a C-CD-F-FG surface). A library design based on Tencon alternative C-CD-F-FG surface is described in U.S. Pat. Publ. No. US2013/0226834. Library designed based on Tencon sequence also includes libraries designed based on Tencon variants, such as Tencon variants having substitutions at residues positions 11, 14, 17, 37, 46, 73, or 86 (residue numbering corresponding to SEQ ID NO: 1), and which variants display improve thermal stability. Exemplary Tencon variants are described in US Pat. Publ. No. 2011/0274623, and include Tencon27 (SEQ ID NO: 4) having substitutions E 11R, L17A, N46V and E861 when compared to Tencon of SEQ ID NO: 1.
TABLE 1
Tencon
FN3 domain (SEQ ID NO: 1)
A strand  1-12
AB loop 13-16
B strand 17-21
BC loop 22-28
C strand 29-37
CD loop 38-43
D strand 44-50
DE loop 51-54
E strand 55-59
EF loop 60-64
F strand 65-74
FG loop 75-81
G strand 82-89
Tencon and other FN3 sequence based libraries may be randomized at chosen residue positions using a random or defined set of amino acids. For example, variants in the library having random substitutions may be generated using NNK codons, which encode all 20 naturally occurring amino acids. In other diversification schemes, DVK codons may be used to encode amino acids Ala, Trp, Tyr, Lys, Thr, Asn, Lys, Ser, Arg, Asp, Glu, Gly, and Cys. Alternatively, NNS codons may be used to give rise to all 20 amino acid residues and simultaneously reducing the frequency of stop codons. Libraries of FN3 domains with biased amino acid distribution at positions to be diversified may be synthesized for example using Slonomics® technology (http:_/www_sloning_com). This technology uses a library of pre-made double stranded triplets that act as universal building blocks sufficient for thousands of gene synthesis processes. The triplet library represents all possible sequence combinations necessary to build any desired DNA molecule. The codon designations are according to the well-known IUB code.
The FN3 domains that specifically bind PD-L1 of the invention may be isolated by producing the FN3 library such as the Tencon library using cis display to ligate DNA fragments encoding the scaffold proteins to a DNA fragment encoding RepA to generate a pool of protein-DNA complexes formed after in vitro translation wherein each protein is stably associated with the DNA that encodes it (U.S. Pat. No. 7,842,476; Odegrip et al., Proc Natl Acad Sci U S A 101, 2806-2810, 2004), and assaying the library for specific binding to PSMA by any method known in the art and described in the Example. Exemplary well known methods which can be used are ELISA, sandwich immunoassays, and competitive and non-competitive assays (see, e.g., Ausubel et al., eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York). The identified FN3 domains that specifically bind PD-L1 are further characterized for their binding to PD-L1, modulation of PD-L1 activity, internalization, stability, and other desired characteristics.
The FN3 domains that specifically bind PD-L1 of the invention may be generated using any FN3 domain as a template to generate a library and screening the library for molecules specifically binding PD-L1 using methods provided within. Exemplar FN3 domains that may be used are the 3rd FN3 domain of tenascin C (TN3) (SEQ ID NO: 125), Fibcon (SEQ ID NO: 126), and the 10th FN3 domain of fibronectin (FN10) (SEQ ID NO: 127). Standard cloning and expression techniques are used to clone the libraries into a vector or synthesize double stranded cDNA cassettes of the library, to express, or to translate the libraries in vitro. For example ribosome display (Hanes and Pluckthun, Proc Natl Acad Sci USA, 94, 4937-4942, 1997), mRNA display (Roberts and Szostak, Proc Natl Acad Sci USA, 94, 12297-12302, 1997), or other cell-free systems (U.S. Pat. No. 5,643,768) can be used. The libraries of the FN3 domain variants may be expressed as fusion proteins displayed on the surface for example of any suitable bacteriophage. Methods for displaying fusion polypeptides on the surface of a bacteriophage are well known (U. S. Pat. Publ. No. 2011/0118144; Int. Pat. Publ. No. WO2009/085462; U.S. Pat. No. 6,969,108; U.S. Pat. No. 6,172,197; U.S. Pat. No. 5,223,409; U.S. Pat. No. 6,582,915; U.S. Pat. No. 6,472,147).
In some embodiments, the FN3 domain that specifically binds PD-L1 is based on Tencon sequence of SEQ ID NO: 1 or Tencon27 sequence of SEQ ID NO: 4, the SEQ ID NO: 1 or the SEQ ID NO: 4, optionally having substitutions at residues positions 11, 14, 17, 37, 46, 73, and/or 86.
The FN3 domains that specifically bind PD-L1 of the invention may be modified to improve their properties such as improve thermal stability and reversibility of thermal folding and unfolding. Several methods have been applied to increase the apparent thermal stability of proteins and enzymes, including rational design based on comparison to highly similar thermostable sequences, design of stabilizing disulfide bridges, mutations to increase alpha-helix propensity, engineering of salt bridges, alteration of the surface charge of the protein, directed evolution, and composition of consensus sequences (Lehmann and Wyss, Curr Opin Biotechnol, 12, 371-375, 2001). High thermal stability may increase the yield of the expressed protein, improve solubility or activity, decrease immunogenicity, and minimize the need of a cold chain in manufacturing. Residues that may be substituted to improve thermal stability of Tencon (SEQ ID NO: 1) are residue positions 11, 14, 17, 37, 46, 73, or 86, and are described in US Pat. Publ. No. 2011/0274623. Substitutions corresponding to these residues may be incorporated to the FN3 domain containing molecules of the invention.
Measurement of protein stability and protein lability can be viewed as the same or different aspects of protein integrity. Proteins are sensitive or “labile” to denaturation caused by heat, by ultraviolet or ionizing radiation, changes in the ambient osmolarity and pH if in liquid solution, mechanical shear force imposed by small pore-size filtration, ultraviolet radiation, ionizing radiation, such as by gamma irradiation, chemical or heat dehydration, or any other action or force that may cause protein structure disruption. The stability of the molecule can be determined using standard methods. For example, the stability of a molecule can be determined by measuring the thermal melting (“Tm”) temperature, the temperature in ° Celsius (° C.) at which half of the molecules become unfolded, using standard methods. Typically, the higher the Tm, the more stable the molecule. In addition to heat, the chemical environment also changes the ability of the protein to maintain a particular three dimensional structure.
In one embodiment, the FN3 domain that specifically binds PD-L1 of the invention may exhibit increased stability by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% or more compared to the same domain prior to engineering measured by the increase in the T..
Chemical denaturation can likewise be measured by a variety of methods. Chemical denaturants include guanidinium hydrochloride, guanidinium thiocyanate, urea, acetone, organic solvents (DMF, benzene, acetonitrile), salts (ammonium sulfate, lithium bromide, lithium chloride, sodium bromide, calcium chloride, sodium chloride); reducing agents (e.g. dithiothreitol, beta-mercaptoethanol, dinitrothiobenzene, and hydrides, such as sodium borohydride), non-ionic and ionic detergents, acids (e.g. hydrochloric acid (HCl), acetic acid (CH3COOH), halogenated acetic acids), hydrophobic molecules (e.g. phosopholipids), and targeted denaturants. Quantitation of the extent of denaturation can rely on loss of a functional property, such as ability to bind a target molecule, or by physiochemical properties, such as tendency to aggregation, exposure of formerly solvent inaccessible residues, or disruption or formation of disulfide bonds.
The FN3 domain that specifically binds PD-L1 of the invention may be generated as monomers, dimers, or multimers, for example, as a means to increase the valency and thus the avidity of target molecule binding, or to generate bi- or multispecific scaffolds simultaneously binding two or more different target molecules. The dimers and multimers may be generated by linking monospecific, bi- or multispecific protein scaffolds, for example, by the inclusion of an amino acid linker, for example a linker containing poly-glycine, glycine and serine, or alanine and proline. Exemplary linker include (GS)2, (SEQ ID NO: 128), (GGGS)2 (SEQ ID NO: 129), (GGGGS)5 (SEQ ID NO: 130), (AP)2 (SEQ ID NO: 131), (AP)5 (SEQ ID NO: 132), (AP)10 (SEQ ID NO: 133), (AP)20 (SEQ ID NO: 134) and A(EAAAK)5AAA (SEQ ID NO: 135). The dimers and multimers may be linked to each other in a N-to C-direction. The use of naturally occurring as well as artificial peptide linkers to connect polypeptides into novel linked fusion polypeptides is well known in the literature (Hallewell et al., J Biol Chem 264, 5260-5268, 1989; Alfthan et al., Protein Eng. 8, 725-731, 1995; Robinson & Sauer, Biochemistry 35, 109-116, 1996; U.S. Pat. No. 5,856,456).
Half-Life Extending Moieties
The FN3 domains that specifically bind PD-L1 of the invention may incorporate other subunits for example via covalent interaction. In one aspect of the invention, the FN3 domains that specifically bind PD-L1 of the invention further comprise a half-life extending moiety. Exemplary half-life extending moieties are albumin, albumin variants, albumin-binding proteins and/or domains, transferrin and fragments and analogues thereof, and Fc regions. An exemplary albumin variant is shown in SEQ ID NO: 136. Amino acid sequences of the human Fc regions are well known, and include IgG1, IgG2, IgG3, IgG4, IgM, IgA and IgE Fc regions.
All or a portion of an antibody constant region may be attached to the FN3 domain that specifically binds PD-L1 of the invention to impart antibody-like properties, especially those properties associated with the Fc region, such as Fc effector functions such as Clq binding, complement dependent cytotoxicity (CDC), Fc receptor binding, antibody-dependent cell-mediated cytotoxicity (ADCC), phagocytosis, down regulation of cell surface receptors (e.g., B cell receptor; BCR), and may be further modified by modifying residues in the Fc responsible for these activities (for review; see Strohl, Curr Opin Biotechnol. 20, 685-691, 2009).
Additional moieties may be incorporated into the FN3 domains that specifically bind PD-L1 of the invention such as polyethylene glycol (PEG) molecules, such as PEG5000 or PEG20,000, fatty acids and fatty acid esters of different chain lengths, for example laurate, myristate, stearate, arachidate, behenate, oleate, arachidonate, octanedioic acid, tetradecanedioic acid, octadecanedioic acid, docosanedioic acid, and the like, polylysine, octane, carbohydrates (dextran, cellulose, oligo- or polysaccharides) for desired properties. These moieties may be direct fusions with the protein scaffold coding sequences and may be generated by standard cloning and expression techniques. Alternatively, well known chemical coupling methods may be used to attach the moieties to recombinantly produced molecules of the invention.
A pegyl moiety may for example be added to the FN3 domain that specifically binds PD-L1 of the invention by incorporating a cysteine residue to the C-terminus of the molecule, or engineering cysteines into residue positions that face away from the PD-L1 binding face of the molecule, and attaching a pegyl group to the cysteine using well known methods.
FN3 domains that specifically bind PD-L1 of the invention incorporating additional moieties may be compared for functionality by several well-known assays. For example, altered properties due to incorporation of Fc domains and/or Fc domain variants may be assayed in Fc receptor binding assays using soluble forms of the receptors, such as the FcγRI, FcγRII, FcγRIII or FcRn receptors, or using well known cell-based assays measuring for example ADCC or CDC, or evaluating pharmacokinetic properties of the molecules of the invention in in vivo models.
Polynucleotides, Vectors, Host Cells
The invention also provides nucleic acids encoding the FN3 domains specifically binding PD-L1 of the invention as isolated polynucleotides or as portions of expression vectors or as portions of linear DNA sequences, including linear DNA sequences used for in vitro transcription/translation, vectors compatible with prokaryotic, eukaryotic or filamentous phage expression, secretion and/or display of the compositions or directed mutagens thereof. Certain exemplary polynucleotides are disclosed herein, however, other polynucleotides which, given the degeneracy of the genetic code or codon preferences in a given expression system, encode the FN3 domains of the invention are also within the scope of the invention.
The invention also provides an isolated polynucleotide encoding the FN3 domain specifically binding PD-L1 comprising the amino acid sequence of SEQ ID NOs: 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123 or 124.
The polynucleotides of the invention may be produced by chemical synthesis such as solid phase polynucleotide synthesis on an automated polynucleotide synthesizer and assembled into complete single or double stranded molecules. Alternatively, the polynucleotides of the invention may be produced by other techniques such as PCR followed by routine cloning. Techniques for producing or obtaining polynucleotides of a given known sequence are well known in the art.
The polynucleotides of the invention may comprise at least one non-coding sequence, such as a promoter or enhancer sequence, intron, polyadenylation signal, a cis sequence facilitating RepA binding, and the like. The polynucleotide sequences may also comprise additional sequences encoding additional amino acids that encode for example a marker or a tag sequence such as a histidine tag or an HA tag to facilitate purification or detection of the protein, a signal sequence, a fusion protein partner such as RepA, Fc or bacteriophage coat protein such as pIX or pIII.
The invention also provides a vector comprising at least one polynucleotide of the invention. Such vectors may be plasmid vectors, viral vectors, vectors for baculovirus expression, transposon based vectors or any other vector suitable for introduction of the polynucleotides of the invention into a given organism or genetic background by any means. Such vectors may be expression vectors comprising nucleic acid sequence elements that can control, regulate, cause or permit expression of a polypeptide encoded by such a vector. Such elements may comprise transcriptional enhancer binding sites, RNA polymerase initiation sites, ribosome binding sites, and other sites that facilitate the expression of encoded polypeptides in a given expression system. Such expression systems may be cell-based, or cell-free systems well known in the art.
The invention also provides a host cell comprising the vector of the invention. The FN3 domain that specifically bind PD-L1 of the invention may be optionally produced by a cell line, a mixed cell line, an immortalized cell or clonal population of immortalized cells, as well known in the art. See, e.g., Ausubel, et al., ed., Current Protocols in Molecular Biology, John Wiley & Sons, Inc., NY, N.Y. (1987-2001); Sambrook, et al., Molecular Cloning: A Laboratory Manual, rd Edition, Cold Spring Harbor, N.Y. (1989); Harlow and Lane, Antibodies, a Laboratory Manual, Cold Spring Harbor, N.Y. (1989); Colligan, et al., eds., Current Protocols in Immunology, John Wiley & Sons, Inc., NY (1994-2001); Colligan et al., Current Protocols in Protein Science, John Wiley & Sons, NY, NY, (1997-2001).
The host cell chosen for expression may be of mammalian origin or may be selected from COS-1, COS-7, HEK293, BHK21, CHO, BSC-1, He G2, SP2/0, HeLa, myeloma, lymphoma, yeast, insect or plant cells, or any derivative, immortalized or transformed cell thereof. Alternatively, the host cell may be selected from a species or organism incapable of glycosylating polypeptides, e.g. a prokaryotic cell or organism, such as BL21, BL21(DE3), BL21-GOLD(DE3), XL1-Blue, JM109, HMS174, HMS174(DE3), and any of the natural or engineered E. coli spp, Klebsiella spp., or Pseudomonas spp strains.
The invention also provides a method of producing the isolated FN3 domain that specifically binds PD-L1 of the invention, comprising culturing the isolated host cell of the invention under conditions such that the isolated FN3 domain that specifically binds PD-L1 is expressed, and purifying the FN3 domain.
The FN3 domains that specifically bind PD-L1 may be purified from recombinant cell cultures by well-known methods, for example by protein A purification, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography, or high performance liquid chromatography (HPLC).
Anti-Idiotypic Antibodies
The present invention also provides an anti-idiotypic antibody binding to the FN3 domain of the invention.
The invention also provides an anti-idiotypic antibody that specifically binds the FN3 domain comprising any one of SEQ ID NOs: 34-124.
Kits
The invention also provides a kit comprising the FN3 domain that specifically binds PD-L1 of the invention.
The kit may be used for therapeutic uses and as a diagnostic kit.
In some embodiments, the kit comprises the FN3 domain that specifically binds PD-L1 of the invention and reagents for detecting the FN3 domain. The kit can include one or more other elements including: instructions for use; other reagents, e.g., a label, an agent useful for chelating, or otherwise coupling, a radioprotective composition; devices or other materials for preparing the FN3 domain that specifically binds PD-L1 of the invention for administration for imaging, diagnostic or therapeutic purpose; pharmaceutically acceptable carriers; and devices or other materials for administration to a subject.
In some embodiments, the kit comprises the FN3 domain that specifically binds PD-L1 comprising any one of SEQ ID NOs: 34-124.
Uses of PD-L1 Binding FN3 Domains of the Invention
The FN3 domains that specifically bind PD-L1 of the invention may be used to diagnose, monitor, modulate, treat, alleviate, help prevent the incidence of, or reduce the symptoms of human disease or specific pathologies in cells, tissues, organs, fluid, or, generally, a host. The FN3 domains that specifically bind PD-L1 of the invention may also be used in imaging PD-L1 positive tumor tissue in a subject. The methods of the invention may be used with an animal patient belonging to any classification. Examples of such animals include mammals such as humans, rodents, dogs, cats and farm animals.
The invention provides a method of diagnosing a subject having, or who is likely to develop cancer of a tissue based on the expression of PD-L1 by cells of the cancer tissue, methods of predicting success of immunotherapy, methods of prognosis, and methods of treatment.
The invention also provides a method of detecting PD-L1-expressing cancer cells in a tumor tissue, comprising
obtaining a sample of the tumor tissue from a subject;
detecting whether PD-L1 is expressed in the tumor tissue by contacting toe sample of the tumor tissues with the FN3 domain that specifically binds PD-L1 comprising the sequence of any one of SEQ ID NOs: 34-124 and detecting the binding between PD-L1 and the FN3 domain.
The tissue can be tissue of any organ or anatomical system, for example lung, epithelial, connective, vascular, muscle, neural, skeletal, lymphatic, prostate, cervical, breast, spleen, gastric, intestinal, oral, esophageal, uterine, ovarian, renal or testicular tissue.
PD-L1 expression may be evaluated using known methods such as immunohistochemistry or ELISA.
The invention also provides a method of isolating PD-L1 expressing cells, comprising
obtaining a sample from a subject;
contacting the sample with the FN3 domain that specifically binds PD-L1 comprising the sequence of any one of SEQ ID NOs: 34-124, and
isolating the cells bound to the FN3 domains.
The invention also provides a method of detecting PD-L1-expressing cancer cells in a tumor tissue, comprising
    • conjugating the FN3 domain that specifically binds PD-L1 comprising the
    • sequence of any one of SEQ ID NOs: 34-124 to a detectable label to form a conjugate;
    • administering the conjugate to a subject; and
    • visualizing the PD-L1 expressing cancer cells to which the conjugate is bound.
The invention also provides a method of treating a subject having cancer, comprising administering to the subject a FN3 domain that specifically binds PD-L1 of the invention.
In some embodiments, the subject has a solid tumor.
In some embodiments, the subject has a hematological malignancy.
In some embodiments, the solid tumor is a melanoma.
In some embodiments, the solid tumor is a lung cancer.
In some embodiments, the solid tumor is a non-small cell lung cancer (NSCLC).
In some embodiments, the solid tumor is a squamous non-small cell lung cancer (NSCLC).
In some embodiments, the solid tumor is a non-squamous NSCLC.
In some embodiments, the solid tumor is a lung adenocarcinoma.
In some embodiments, the solid tumor is a renal cell carcinoma (RCC).
In some embodiments, the solid tumor is a mesothelioma.
In some embodiments, the solid tumor is a nasopharyngeal carcinoma (NPC).
In some embodiments, the solid tumor is a colorectal cancer.
In some embodiments, the solid tumor is a prostate cancer.
In some embodiments, the solid tumor is castration-resistant prostate cancer.
In some embodiments, the solid tumor is a stomach cancer.
In some embodiments, the solid tumor is an ovarian cancer.
In some embodiments, the solid tumor is a gastric cancer.
In some embodiments, the solid tumor is a liver cancer.
In some embodiments, the solid tumor is pancreatic cancer.
In some embodiments, the solid tumor is a thyroid cancer.
In some embodiments, the solid tumor is a squamous cell carcinoma of the head and neck.
In some embodiments, the solid tumor is a carcinomas of the esophagus or gastrointestinal tract.
In some embodiments, the solid tumor is a breast cancer.
In some embodiments, the solid tumor is a fallopian tube cancer.
In some embodiments, the solid tumor is a brain cancer.
In some embodiments, the solid tumor is an urethral cancer.
In some embodiments, the solid tumor is a genitourinary cancer.
In some embodiments, the solid tumor is an endometriosis.
In some embodiments, the solid tumor is a cervical cancer.
In some embodiments, the solid tumor is a metastatic lesion of the cancer.
In some embodiments, the hematological malignancy is a lymphoma, a myeloma or a leukemia.
In some embodiments, the hematological malignancy is a B cell lymphoma.
In some embodiments, the hematological malignancy is Burkitt's lymphoma.
In some embodiments, the hematological malignancy is Hodgkin's lymphoma.
In some embodiments, the hematological malignancy is a non-Hodgkin's lymphoma.
In some embodiments, the hematological malignancy is a myelodysplastic syndrome.
In some embodiments, the hematological malignancy is an acute myeloid leukemia (AML).
In some embodiments, the hematological malignancy is a chronic myeloid leukemia (CML).
In some embodiments, the hematological malignancy is a chronic myelomoncytic leukemia (CMML).
In some embodiments, the hematological malignancy is a multiple myeloma (MM).
In some embodiments, the hematological malignancy is a plasmacytoma. In some embodiments, the cancer is kidney cancer.
“Treat” or “treatment” refers to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) an undesired physiological change or disorder, such as the development or spread of cancer. For purposes of this invention, beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. “Treatment” can also mean prolonging survival as compared to expected survival if not receiving treatment. Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented.
A “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve a desired therapeutic result. A therapeutically effective amount of the FN3 domains that specifically bind PD-L1 of the invention may vary according to factors such as the disease state, age, sex, and weight of the individual. Exemplary indicators of an effective FN3 domain that specifically binds PD-L1 is improved well-being of the patient, decrease or shrinkage of the size of a tumor, arrested or slowed growth of a tumor, and/or absence of metastasis of cancer cells to other locations in the body.
Administration/Pharmaceutical Compositions
The invention provides for pharmaceutical compositions of the FN3 domains that specifically bind PD-L1, optionally conjugated to a detectable label or a cytotoxic drug of the invention and a pharmaceutically acceptable carrier. For therapeutic use, the FN3 domains that specifically bind PD-L1 of the invention may be prepared as pharmaceutical compositions containing an effective amount of the domain or molecule as an active ingredient in a pharmaceutically acceptable carrier. “Carrier” refers to a diluent, adjuvant, excipient, or vehicle with which the active compound is administered. Such vehicles can be liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. For example, 0.4% saline and 0.3% glycine can be used. These solutions are sterile and generally free of particulate matter. They may be sterilized by conventional, well-known sterilization techniques (e.g., filtration). The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, stabilizing, thickening, lubricating and coloring agents, etc. The concentration of the molecules of the invention in such pharmaceutical formulation can vary widely, i.e., from less than about 0.5%, usually at least about 1% to as much as 15 or 20% by weight and will be selected primarily based on required dose, fluid volumes, viscosities, etc., according to the particular mode of administration selected. Suitable vehicles and formulations, inclusive of other human proteins, e.g., human serum albumin, are described, for example, in e.g. Remington: The Science and Practice of Pharmacy, 21′ Edition, Troy, D.B. ed., Lipincott Williams and Wilkins, Philadelphia, PA 2006, Part 5, Pharmaceutical Manufacturing pp 691-1092, See especially pp. 958-989.
The mode of administration for therapeutic use of the FN3 domains of the invention may be any suitable route that delivers the agent to the host, such as parenteral administration, e.g., intradermal, intramuscular, intraperitoneal, intravenous or subcutaneous, pulmonary; transmucosal (oral, intranasal, intravaginal, rectal), using a formulation in a tablet, capsule, solution, powder, gel, particle; and contained in a syringe, an implanted device, osmotic pump, cartridge, micropump; or other means appreciated by the skilled artisan, as well known in the art. Site specific administration may be achieved by for example intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracerebellar, intracerebroventricular, intracolic, intracervical, intragastric, intrahepatic, intracardial, intraosteal, intrapelvic, intrapericardiac, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravascular, intravesical, intralesional, vaginal, rectal, buccal, sublingual, intranasal, or transdermal delivery.
Pharmaceutical compositions can be supplied as a kit comprising a container that comprises the pharmaceutical composition as described herein. A pharmaceutical composition can be provided, for example, in the form of an injectable solution for single or multiple doses, or as a sterile powder that will be reconstituted before injection. Alternatively, such a kit can include a dry-powder disperser, liquid aerosol generator, or nebulizer for administration of a pharmaceutical composition. Such a kit can further comprise written information on indications and usage of the pharmaceutical composition.
While having described the invention in general terms, the embodiments of the invention will be further disclosed in the following examples that should not be construed as limiting the scope of the claims.
EXAMPLE 1 Construction of Tencon Libraries with Randomized Loops
Tencon (SEQ ID NO: 1) is an immunoglobulin-like scaffold, fibronectin type III (FN3) domain, designed from a consensus sequence of fifteen FN3 domains from human tenascin-C (Jacobs et al., Protein Engineering, Design, and Selection, 25:107-117, 2012; U.S. Pat. No. 8,278,419). The crystal structure of Tencon shows six surface-exposed loops that connect seven beta-strands. These loops, or selected residues within each loop, can be randomized in order to construct libraries of fibronectin type III (FN3) domains that can be used to select novel molecules that bind to specific targets.
Tencon:
(SEQ ID NO 1)
LPAPKNLVVSEVTEDSLRLSWTAPDAAFDSFLIQYQESEKVGEAINLTVP
GSERSYDLTGLKPGTEYTVSIYGVKGGHRSNPLSAEFTT:

Various libraries were generated using the tencon scaffold and various design strategies. In general, libraries TCL1 and TCL2 produced good binders. Generation of TCL1 and TCL2 libraries are described in detail in Int. Pat. Publ. No. WO/2014081944A2.
Construction of TCL1 Library
A library designed to randomize only the FG loop of Tencon (SEQ ID NO: 1), TCL1, was constructed for use with the cis-display system (Jacobs et al., Protein Engineering, Design, and Selection, 25:107-117, 2012). In this system, a single-strand DNA incorporating sequences for a Tac promoter, Tencon library coding sequence, RepA coding sequence, cis-element, and ori element is produced. Upon expression in an in vitro transcription/translation system, a complex is produced of the Tencon-RepA fusion protein bound in cis to the DNA from which it is encoded. Complexes that bind to a target molecule are then isolated and amplified by polymerase chain reaction (PCR), as described below.
Construction of the TCL1 library for use with cis-display was achieved by successive rounds of PCR to produce the final linear, double-stranded DNA molecules in two halves; the 5′ fragment contains the promoter and Tencon sequences, while the 3′ fragment contains the repA gene and the cis- and ori elements. These two halves are combined by restriction digest in order to produce the entire construct. The TCL1 library was designed to incorporate random amino acids only in the FG loop of Tencon, KGGHRSN (SEQ ID NO: 55). NNS codons were used in the construction of this library, resulting in the possible incorporation of all 20 amino acids and one stop codon into the FG loop. The TCL1 library contains six separate sub-libraries, each having a different randomized FG loop length, from 7 to 12 residues, in order to further increase diversity.
TCL1 library
(SEQ ID NO: 2)
LPAPKNLVVSEVTEDSLRLSWTAPDAAFDSFLIQYQESEKVGEAINLTVP
GSERSYDLTGLKPGTEYTVSIYGVX1X2X3X4X5X6X7X8X9X10X11
X12 PLSAEFTT; 
wherein
X1, X2, X3, X4, X5, X6, X7 is any amino acid; and
X8, X9, X10, X11 and X12 are any amino acid or
deleted

Construction of TCL2 Library
TCL2 library was constructed in which both the BC and the FG loops of Tencon were randomized and the distribution of amino acids at each position was strictly controlled. Table 3 shows the amino acid distribution at desired loop positions in the TCL2 library. The designed amino acid distribution had two aims. First, the library was biased toward residues that were predicted to be structurally important for Tencon folding and stability based on analysis of the Tencon crystal structure and/or from homology modeling. For example, position 29 was fixed to be only a subset of hydrophobic amino acids, as this residue was buried in the hydrophobic core of the Tencon fold. A second layer of design included biasing the amino acid distribution toward that of residues preferentially found in the heavy chain HCDR3 of antibodies, to efficiently produce high-affinity binders (Birtalan et al., J Mol Biol 377:1518-28, 2008; Olson et al., Protein Sci 16:476-84, 2007). Towards this goal, the “designed distribution” in Table 2 refers to the distribution as follows: 6% alanine, 6% arginine, 3.9% asparagine, 7.5% aspartic acid, 2.5% glutamic acid, 1.5% glutamine, 15% glycine, 2.3% histidine, 2.5% isoleucine, 5% leucine, 1.5% lysine, 2.5% phenylalanine, 4% proline, 10% serine, 4.5% threonine, 4% tryptophan, 17.3% tyrosine, and 4% valine. This distribution is devoid of methionine, cysteine, and STOP codons.
TCL2 library
(SEQ ID NO: 3)
LPAPKNLVVSEVTEDSLRLSWX1X2X3X4X5X6X7X8SFLIQYQESEKVG
EAINLTVPGSERSYDLTGLKPGTEYTVSIYGVX9X10X11X12X13SX14
X15LSAEFTT;
wherein
X1 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile,
Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or Val;
X2 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile,
Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or Val;
X3 Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile,
Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or Val;
X4 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile,
Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or Val;
X5 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile,
Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or Val;
X6 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile,
Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or Val;
X7 is Phe, Ile, Leu, Val or Tyr;
X8 is Asp, Glu or Thr;
X9 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile,
Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or Val;
X10 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His,
Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or
Val;
X11 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His,
Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or
Val;
X12 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His,
Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or
Val;
X13 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His,
Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or
Val;
X14 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His,
Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or
Val;
and
X15 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His,
Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or
Val.
TABLE 2
Residue
Position* WT residues Distribution in the TCL2 library
22 T designed distribution
23 A designed distribution
24 P 50% P + designed distribution
25 D designed distribution
26 A 20% A + 20% G + designed distribution
27 A designed distribution
28 F 20% F, 20% I, 20% L, 20% V, 20% Y
29 D 33% D, 33% E, 33% T
75 K designed distribution
76 G designed distribution
77 G designed distribution
78 H designed distribution
79 R designed distribution
80 S 100% S
81 N designed distribution
82 P 50% P + designed distribution
*residue numbering is based on Tencon sequence of SEQ ID NO: 1
Subsequently, these libraries were improved by various ways, including building of the libraries on a stabilized Tencon framework (U.S. Pat. No. 8,569,227) that incorporates substitutions El1R/L17A/N46V/E861 (Tencon27; SEQ ID NO: 4) when compared to the wild type tencon as well as altering of the positions randomized in the BC and FG loops. Tencon27 is described in Int. Pat. Appl. No. WO2013049275. From this, new libraries designed to randomize only the FG loop of Tencon (library TCL9), or a combination of the BC and FG loops (library TCL7) were generated. These libraries were constructed for use with the cis-display system (Odegrip et al., Proc Natl Acad Sci U S A 101: 2806-2810, 2004). The details of this design are shown below:
Stabilized Tencon (Tencon27)
(SEQ ID NO: 4)
LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFLIQYQESEKVGEAIVLTVP
GSERSYDLTGLKPGTEYTVSIYGVKGGHRSNPLSAIFTT
TCL7 (randomized FG and BC loops)
(SEQ ID NO: 5)
LPAPKNLVVSRVTEDSARLSWX1X2X3X4X5X6X7X8X9FDSFLIQYQES
EKVGEAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVX10X11X12X13
X14X15X16X17X18X19SNPLSAIFTT;
wherein
X1, X2, X3, X4, X5, X6, X10, X11, X12, X13, X14,
X15 and X16 is A, D, E, F, G, H, I, K, L, N, P, Q,
R, S, T, V, W or Y;
and
X7, X8, X9, X17, X18 and X19, is A, D, E, F, G, H,
I, K, L, N, P, Q, R, S, T, V, W, Y or deleted.
TCL9 (randomized FG loop)
(SEQ ID NO: 6)
LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFLIQYQESEKVGEAIVLTVP
GSERSYDLTGLKPGTEYTVSIYGV XX2X3X4X5X6X7X8X9 X10X11
X12SNPLSAIFTT;
X1, X2, X3, X4, X5, X6 and X7, is A, D, E, F, G,
H, I, K, L, N, P, Q, R, S, T, V, W or Y;
and
X8, X9, X10, X11 and X12 is A, D, E, F, G, H, I,
K, L, N, P, Q, R, S, T, V, W, Y or deleted.
For library construction, DNA fragments encoding randomized BC loops (lengths 6-9 positions) or FG loops (lengths 7-12 positions) were synthesized using Slonomics technology (Sloning Biotechnology GmbH) so as to control the amino acid distribution of the library and to eliminate stop codons. Two different sets of DNA molecules randomizing either the BC loop or the FG loops were synthesized independently and later combined using PCR to produce the full library product.
Construction of FG Loop Libraries (TCL9)
A set of synthetic DNA molecules consisting of a 5′ Tac promoter followed by the complete gene sequence of Tencon with the exception of randomized codons in the FG loop was produced (SEQ ID NOs: 26-31). For FG loop randomization, all amino acids except cysteine and methionine were encoded at equal percentages. The lengths of the diversified portion are such that they encode for 7, 8, 9, 10, 11, or 12 amino acids in the FG loop. Sub-libraries of each length variation were synthesized individually at a scale of 2 ug and then amplified by PCR using oligos Sloning-FOR (SEQ ID NO: 9) and Sloning-Rev (SEQ ID NO: 10).
The 3′ fragment of the library is a constant DNA sequence containing elements for display, including a PspOMI restriction site, the coding region of the repA gene, and the cis- and ori elements. PCR reactions were performed to amplify this fragment using a plasmid (pCR4Blunt) (Invitrogen) as a template with M13 Forward and M13 Reverse primers. The resulting PCR products were digested by PspOMI overnight and gel-purified. To ligate the 5′ portion of library DNA to the 3′ DNA containing repA gene, 2 pmol (˜540 ng to 560 ng) of 5′ DNA was ligated to an equal molar (˜1.25 ug) of 3′ repA DNA in the presence of NotI and PspOMI enzyme and T4 ligase at 37° C. overnight. The ligated library product was amplified by using 12 cycles of PCR with oligos POP2250 (SEQ ID NO: 11) and DigLigRev (SEQ ID NO: 12). For each sub-library, the resulting DNA from 12 PCR reactions were combined and purified by Qiagen spin column The yield for each sub-library of TCL9 ranged from 32-34 lag.
Construction of FG/BC Loop Libraries (TCL7)
The TCL7 library provides for a library with randomized Tencon BC and FG loops. In this library, BC loops of lengths 6-9 amino acids were mixed combinatorially with randomized FG loops of 7-12 amino acids in length. Synthetic Tencon fragments BC6, BC7, BC8, and BC9 (SEQ ID NOs: 13-16, respectively) were produced to include the Tencon gene encoding for the N-terminal portion of the protein up to and including residue VX such that the BC loop is replaced with either 6, 7, 8, or 9 randomized amino acids. These fragments were synthesized prior to the discovery of L17A, N46V and E831 mutations (CEN5243) but these mutations were introduced in the molecular biology steps described below. In order to combine this fragment with fragments encoding for randomized FG loops, the following steps were taken.
First, a DNA fragment encoding the Tac promoter and the 5′ sequence of Tencon up to the nucleotide encoding for amino acid A17 (130mer-L17A, SEQ ID NO: 17) was produced by PCR using oligos POP2222ext (SEQ ID NO: 18) and LS1114 (SEQ ID NO: 19). This was done to include the L17A mutation in the library (CEN5243). Next, DNA fragments encoding for Tencon residues R18-V75 including randomized BC loops were amplified by PCR using BC6, BC7, BC8, or BC9 as a templates and oligos LS1115 (SEQ ID NO: 20) and LS1117 (SEQ ID NO: 21). This PCR step introduced a Bsal site at the 3′ end. These DNA fragments were subsequently joined by overlapping PCR using oligos POP2222ext and LS1117 as primers. The resulting PCR product of 240 bp was pooled and purified by Qiagen PCR purification kit. The purified DNA was digested with Bsal-HF and gel purified.
Fragments encoding the FG loop were amplified by PCR using FG7, FG8, FG9, FG10, FG11, and FG12 as templates with oligonucleotides SDG10 (SEQ ID NO: 22) and SDG24 (SEQ ID NO: 23) to incorporate a Bsal restriction site and N46V and E86I variations (CEN5243).
The digested BC fragments and FG fragments were ligated together in a single step using a 3-way ligation. Four ligation reactions in the 16 possible combinations were set up, with each ligation reaction combining two BC loop lengths with 2 FG loop lengths. Each ligation contained—300 ng of total BC fragment and 300 ng of the FG fragment. These 4 ligation pools were then amplified by PCR using oligos POP2222 (SEQ ID NO: 24) and SDG28 SEQ ID N: 25). 7.5 lag of each reaction product were then digested with Notl and cleaned up with a Qiagen PCR purification column. 5.2 lag of this DNA, was ligated to an equal molar amount of RepA DNA fragment (˜14 lig) digested with PspOMI and the product amplified by PCR using oligos POP2222.
EXAMPLE 2 Generation of Tencon Libraries having Alternative Binding Surfaces
The choice of residues to be randomized in a particular library design governs the overall shape of the interaction surface created. X-ray crystallographic analysis of an FN3 domain containing scaffold protein selected to bind maltose binding protein (MBP) from a library in which the BC, DE, and FG loops were randomized was shown to have a largely curved interface that fits into the active site of MBP (Koide et al., Proc Natl Acad Sci U S A 104: 6632-6637, 2007). In contrast, an ankyrin repeat scaffold protein that was selected to bind to MBP was found to have a much more planar interaction surface and to bind to the outer surface of MBP distant from the active (Binz et al., Nat Biotechnol 22: 575-582, 2004). These results suggest that the shape of the binding surface of a scaffold molecule (curved vs. flat) may dictate what target proteins or specific epitopes on those target proteins are able to be bound effectively by the scaffold. Published efforts around engineering protein scaffolds containing FN3 domains for protein binding has relied on engineering adjacent loops for target binding, thus producing curved binding surfaces. This approach may limit the number of targets and epitopes accessible by such scaffolds.
Tencon and other FN3 domains contain two sets of CDR-like loops lying on the opposite faces of the molecule, the first set formed by the BC, DE, and FG loops, and the second set formed by the AB, CD, and EF loops. The two sets of loops are separated by the beta-strands that form the center of the FN3 structure. If the image of the Tencon is rotated by 90 degrees, an alternative surface can be visualized. This slightly concave surface is formed by the CD and FG loops and two antiparallel beta- strands, the C and the F beta-strands, and is herein called the C-CD-F-FG surface. The C-CD-F-FG surface can be used as a template to design libraries of protein scaffold interaction surfaces by randomizing a subset of residues that form the surface. Beta-strands have a repeating structure with the side chain of every other residue exposed to the surface of the protein. Thus, a library can be made by randomizing some or all surface exposed residues in the beta strands. By choosing the appropriate residues in the beta-strands, the inherent stability of the Tencon scaffold should be minimally compromised while providing a unique scaffold surface for interaction with other proteins.
Library TCL14 (SEQ ID NO: 7), was designed into Tencon27 scaffold (SEQ ID NO: 4).
A full description of the methods used to construct this library is described in US. Pat. Publ. No. US2013/0226834.
TCL14 library (SEQ ID NO: 7):
LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFX1IX2YX3EX4X5X6X7GE
AIVLTVPGSERSYDLTGLKPGTEYX8VX9IX10GVKGGX11X12SX13PL
SAIFTT;
wherein
X1, X2, X3, X4, X5, X6, X7, X8, X9, X10, X11, X12
and X13 are A, D, E, F, G, H, I, K, L, N, P,
Q, R, S, T, V, W, Y, C or M.
The two beta strands forming the C-CD-F-FG surface in Tencon27 have a total of 9 surface exposed residues that could be randomized; C-strand: S30, L32, Q34, Q36; F-strand: E66, T68, S70, Y72, and V74, while the CD loop has 6 potential residues: S38, E39, K40, V41, G42, and E43 and the FG loop has 7 potential residues: K75, G76, G77, H78, R79, S80, and N81. Select residues were chosen for inclusion in the TCL14 design due to the larger theoretical size of the library if all 22 residues were randomized.
Thirteen positions in Tencon were chosen for randomizing: L32, Q34 and Q36 in C-strand, S38, E39, K40 and V41 in CD-loop, T68, S70 and Y72 in F-strand, H78, R79, and N81 in FG-loop. In the C and F strands S30 and E66 were not randomized as they lie just beyond the CD and FG loops and do not appear to be as apparently a part of the C-CD-F-FG surface. For the CD loop, G42 and E43 were not randomized as glycine, providing flexibility, can be valuable in loop regions, and E43 lies at the junction of the surface. The FG loop had K75, G76, G77, and S80 excluded. The glycines were excluded for the reasons above while careful inspection of the crystal structures revealed S80 making key contacts with the core to help form the stable FG loop. K75 faces away from the surface of the C-CD-F-FG surface and was a less appealing candidate for randomization. Although the above mentioned residues were not randomized in the original TCL14 design, they could be included in subsequent library designs to provide additional diversity for de novo selection or for example for an affinity maturation library on a select TCL14 target specific hit.
Subsequent to the production of TCL14, 3 additional Tencon libraries of similar design were produced. These two libraries, TCL19, TCL21 and TCL23, are randomized at the same positions as TCL14 (see above) however the distribution of amino acids occurring at these positions is altered (Table 3). TCL19 and TCL21 were designed to include an equal distribution of 18 natural amino acids at every position (5.55% of each), excluding only cysteine and methionine. TCL23 was designed such that each randomized position approximates the amino acid distribution found in the HCDR3 loops of functional antibodies (Birtalan et al., J Mol Biol 377: 1518-1528, 2008) as described in Table 3. As with the TCL21 library, cysteine and methionine were excluded.
A third additional library was built to expand potential target binding surface of the other libraries library. In this library, TCL24, 4 additional Tencon positions were randomized as compared to libraries TCL14, TCL19, TCL21, and TCL23. These positions include N46 and T48 from the D strand and S84 and 186 from the G strand. Positions 46, 48, 84, and 86 were chosen in particular as the side chains of these residues are surface exposed from beta-strands D and G and lie structurally adjacent to the randomized portions of the C and F strand, thus increasing the surface area accessible for binding to target proteins. The amino acid distribution used at each position for TCL24 is identical to that described for TCL19 and TCL21 in Table 3.
TCL24 Library
(SEQ ID NO: 8)
LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFX1IX2YX3EX4X5X6X7GE
AIX8LX9VPGSERSYDLTGLKPGTEYX10VX11IX12GVKGGX13X14S
X15PLX16AX17FTT;
wherein
X1, X2, X3, X4, X5, X6, X10, X11, X12, X13, X14,
X15, X16 and X17 are A, D, E, F, G, H, I, K, L, N,
P, Q, R, S, T, V, Y or W.
TABLE 3
Amino acid frequency (%) at each randomized
position for TCL21, TCL23, and TCL24.
Amino Acid TCL19 TCL21 TCL23 TCL24
Ala 5.6 5.6 6.0 5.6
Arg 5.6 5.6 6.0 5.6
Asn 5.6 5.6 3.9 5.6
Asp 5.6 5.6 7.5 5.6
Cys 0.0 0.0 0.0 0.0
Gln 5.6 5.6 1.5 5.6
Glu 5.6 5.6 2.5 5.6
Gly 5.6 5.6 15.0 5.6
His 5.6 5.6 2.3 5.6
Ile 5.6 5.6 2.5 5.6
Leu 5.6 5.6 5.0 5.6
Lys 5.6 5.6 1.5 5.6
Met 0.0 0.0 0.0 0.0
Phe 5.6 5.6 2.5 5.6
Pro 5.6 5.6 4.0 5.6
Ser 5.6 5.6 10.0 5.6
Thr 5.6 5.6 4.5 5.6
Trp 5.6 5.6 4.0 5.6
Tyr 5.6 5.6 17.3 5.6
Val 5.6 5.6 4.0 5.6

Generation of TCL21, TCL23, and TCL24 Libraries
The TCL21 library was generated using Colibra library technology (Isogenica) in order to control amino acid distributions. TCL19, TCL23, and TCL24 gene fragments were generated using Slonomics technology (Morphosys) to control amino acid distributions. PCR was used to amplify each library following initial synthesis followed by ligation to the gene for RepA in order to be used in selections using the CIS-display system (Odegrip et al., Proc Natl Acad Sci U S A 101: 2806-2810, 2004) as described above for the loop libraries.
EXAMPLE 3 Selection of Fibronectin Type III (FN3) Domains that Bind PD-L1 Panning
FN3 domains specific for human PD-L1 were selected via CIS-Display (Odegrip et al 2004) using recombinant biotinylated PD-L1 (rhPD-L1/Fc Chimera, R&D Systems 156-B7). For in vitro transcription and translation (ITT), 3μg of DNA from libraries TCL18, TCL19, TCL21, TCL23, and TCL24 were incubated at 30° C. with 0.1 mM complete amino acids, 1X S30 premix components, and 15 μL of S30 extract (Isogenica) in a total volume of 50 μL. After 1 hour, 375 μL of blocking solution (2% BSA in PBS, Invitrogen) was added and reactions were incubated on a cold block for 15 minutes. Unbound library members were removed by successive washes with TBST and TBS. After washing, DNA was eluted from the target protein by heating to 75° C. for 10 minutes and amplified by PCR using KOD polymerase for further rounds of panning High affinity binders were isolated by successively lowering the concentration of target PD-L1 during each round from 400 nM to 100 nM and increasing the washing stringency.
Outputs from the fifth round panning were subjected to four additional rounds of off-rate selection. Library transcription and translation was performed as described above after which the ITT reactions were incubated with biotinylated recombinant PD-L1 proteins and captured on neutravidin or streptavidin coated magnetic beads, before being washed in TBST extensively then subsequently washed in 5 μM cold recombinant PD-L1 protein for 1 hour. The biotinylated target antigen concentration was reduced from 25 nM in rounds 6 and 7 to 2.5 nM in rounds 8 and 9.
Following panning, genes encoding the selected FN3 domains were amplified by PCR, subcloned into a pET vector modified to include a ligase independent cloning site, and transformed into BL21 (DE3) (Stratagene) cells for soluble expression in E. coli using standard molecular biology techniques. A gene sequence encoding a C-terminal poly-histidine tag was added to each FN3 domain to enable purification and detection. Cultures were grown to an optical density of 0.6-0.8 in TB medium supplemented with 100 μg/mL carbenicillin in 1 mL 96-well blocks at 37° C. before the addition of IPTG to 1 mM, at which point the temperature was reduced to 30° C. Cells were harvested approximately 16 hours later by centrifugation and frozen at −20° C. Cell lysis was achieved by incubating each pellet in 0.6 mL of BugBuster® HT lysis buffer (Novagen EMD Biosciences) supplemented with 0.2 mg/mL lysozyme with shaking at room temperature for 30 minutes.
Biochemical Screening for FN3 Domains that Bind Recombinant PD-L1
Streptavidin-coated Maxisorp plates (Nunc catalog 436110) were blocked for lh in Starting Block T20 (Pierce) and then coated with biotinylated PD-L1 (using same antigen as in panning) or negative controls (an unrelated Fc-fused recombinant protein and human serum albumin) for lh. Plates were rinsed with TBST and diluted lysate was applied to plates for 1h. Following additional rinses, wells were treated with HRP-conjugated anti-FN3 domain antibody (PAB25) for lh and then assayed with POD (Roche catalog 11582950001). The DNA from FN3 domain lysates with ELISA binding signals to PD-L1 at least 5-fold above both Fc and HSA controls were sequenced resulting in 57 (Table 4) and 37 (Table 5) unique, readable FN3 domain sequences isolated from Round 5 and Round 9 screening respectively.
High-throughput Expression of Anti-PD-L1 FN3 Domains
40 isolated clones from unique hits identified by biochemical binding ELISA from Round 9 were combined for growth into 96-well block plate; clones grew in 1 mL cultures (LB media supplemented with kanamycin for selection) at 37° C. overnight with shaking. For protein expression in 96-block plates, 1 mL TB media supplemented with kanamycin was inoculated with 50 μL of the overnight culture and grown at 37° C. with continual shaking at 300rpm until OD600 =0.6-1. Once the target OD was reached, protein expression was induced with addition of IPTG to 1 mM; plates were transferred to 30° C. (300 rpm) for overnight growth. Overnight cultures were centrifuged to harvest the cells; bacterial pellets were stored at −80° C. until ready for use. Pellets were lysed with BugBuster® HT lysis buffer (Novagen EMD Biosciences) and His-tagged Centyrins purified from the clarified lysates with His MultiTrap™ HP plates (GE Healthcare) and eluted in buffer containing 20 mM sodium phosphate, 500 mM sodium chloride, and 250 mM imidazole at pH 7.4. Purified samples were exchanged into PBS pH 7.4 for analysis using PD MultiTrap™ G-25 plates (GE Healthcare).
Size Exclusion Chromatography Analysis
Size exclusion chromatography was used to determine the aggregation state of anti-PD-L1 FN3 domains Aliquots (10)(L) of each purified FN3 domain were injected onto a Superdex 75 5/150 column (GE Healthcare) at a flow rate of 0.3 mL/min in a mobile phase of PBS pH 7.4. Elution from the column was monitored by absorbance at 280 nm. Tencon protein was included in each run as a control. Agilent ChemStation software was used to analyse the elution profiles. 20 anti-PD-L1 FN3 domains demonstrated a retention time between 5.2 and 6.4 minutes and only a single SEC peak indicative of monomeric protein (Table 6).
TABLE 4
ELISA ELISA Fc ELISA SEQ
PD-L1 Fc Control HSA ID
Clone (RSU) (RSU) (RSU) NO:
ISOP121HR5P1G9 17760 880 1760 34
ISOP121BR5P1F7 12880 720 880 35
ISOP121BR5P1A6 10960 720 720 36
ISOP121BR5P1C5 11680 400 720 37
ISOP121BR5P1D7 12800 800 720 38
ISOP121BR5P1C6 13360 720 720 39
ISOP121AR5P1G6 16960 1200 880 40
ISOP121BR5P1B7 11360 640 480 41
ISOP121FR5P1G1 10000 640 400 42
ISOP121GR5P1B4 16160 800 560 43
ISOP121BR5P1G2 16720 800 560 44
ISOP121HR5P1H2 20960 720 560 45
ISOP121FR5P1G11 18560 880 480 46
ISOP121AR5P1E7 327200 4240 6560 47
ISOP121GR5P1F6 32080 640 640 48
ISOP121BR5P1E9 42000 960 800 49
ISOP121AR5P1F2 51040 880 960 50
ISOP121AR5P1F7 64000 720 1040 51
ISOP121BR5P1H6 74640 1440 1040 52
ISOP121GR5P1A2 61680 720 720 53
ISOP121BR5P1D3 75760 800 800 54
ISOP121AR5P1F9 136080 1120 1040 55
ISOP121AR5P1H5 170800 960 1120 56
ISOP121AR5P1G10 231920 1360 1280 57
ISOP121AR5P1F3 180160 800 960 58
ISOP121BR5P1E2 137280 800 720 59
ISOP121BR5P1D1 186240 1040 960 60
ISOP121BR5P1C9 226400 1120 1040 61
ISOP121GR5P1G11 239600 960 1040 62
ISOP121BR5P1A7 388640 800 1120 63
ISOP121BR5P1C3 177040 640 480 64
ISOP121AR5P1D11 392800 640 1040 65
ISOP121ER5P1E7 251120 480 560 66
ISOP121GR5P1G7 367760 800 800 67
ISOP121AR5P1A8 515920 560 1040 68
ISOP121BR5P1E7 411760 800 640 69
ISOP121FR5P1H8 430640 560 640 70
ISOP121GR5P1D2 513280 720 640 71
ISOP121AR5P1H2 926720 880 1120 72
ISOP121GR5P1F10 577120 640 640 73
ISOP121BR5P1A2 742800 720 800 74
ISOP121GR5P1F7 697200 640 720 75
ISOP121AR5P1B8 591600 640 560 76
ISOP121GR5P1D7 791920 720 720 77
ISOP121BR5P1G3 770800 560 640 78
ISOP121AR5P1C5 732480 640 560 79
ISOP121FR5P1H9 1195520 720 880 80
ISOP121AR5P1A10 788560 1120 560 81
ISOP121HR5P1F2 906960 480 640 82
ISOP121AR5P1H1 1475280 880 880 83
ISOP121BR5P1D10 1538800 480 880 84
ISOP121BR5P1F10 1422880 560 720 85
ISOP121BR5P1D11 2442960 800 1120 86
ISOP121AR5P1E11 1842000 720 720 87
ISOP121BR5P1D6 2435760 560 880 88
ISOP121BR5P1B5 1483520 720 400 89
TABLE 5
ELISA ELISA Fc ELISA SEQ
PD-L1 Fc Control HSA ID
Clone (RSU) (RSU) (RSU) NO:
ISOP194ER9P1G3 4288320 560 720 90
ISOP194AR9P1F2 16271040 1920 7520 91
ISOP194AR9P1H10 5212800 4400 2400 92
ISOP194BR9P1H4 4064960 3040 3840 93
ISOP194AR9P1D8 923200 12000 6560 94
ISOP194BR9P1D1 2152080 1360 1280 95
ISOP194AR9P1E8 3404480 6960 67680 96
ISOP194AR9P1E9 19719920 5520 1600 97
ISOP194AR9P1H9 2592720 21280 11360 98
ISOP194BR9P1A9 19046640 2320 3200 99
ISOP194BR9P1A5 3182000 800 1280 100
ISOP194BR9P1F7 15151120 1920 1760 101
ISOP194AR9P1G7 15914000 1280 560 102
ISOP194AR9P1E3 4566880 1120 800 103
ISOP194AR9P1C5 4371120 3440 1040 104
ISOP194AR9P1H3 17746800 9200 4880 105
ISOP194GR9P1E9 2821920 720 1200 106
ISOP194HR9P1B10 385360 560 1840 107
ISOP194ER9P1A11 4352240 800 880 108
ISOP194ER9P1A3 2360160 560 800 109
ISOP194ER9P1H9 3042800 720 880 110
ISOP194HR9P1B2 5656400 400 1840 111
ISOP194HR9P1D11 6620160 480 1680 112
ISOP194GR9P1F6 319200 400 1200 113
ISOP194GR9P1F9 105280 320 800 114
ISOP194GR9P1C11 164320 1040 1440 115
ISOP194ER9P1E6 8982160 240 720 116
ISOP194BR9P1G9 14376560 640 960 117
ISOP194BR9P1E4 9791680 640 1440 118
ISOP194AR9P1H1 21445040 15680 6800 119
ISOP194BR9P1D10 1666880 720 1120 120
ISOP194BR9P1C8 6110640 640 1280 121
ISOP194AR9P1C10 13863040 38240 14960 122
ISOP194AR9P1D11 1043280 28160 12720 123
ISOP194AR9P1C3 3548240 56400 5920 124
TABLE 6
SEC SEC Peak SEQ
Retention Height ID
Clone Time (min) (mAU) Monomeric? NO:
ISOP194ER9P1G3 5.951 167.47 FALSE 90
ISOP194AR9P1F2 5.901 552.30 TRUE 91
ISOP194AR9P1H10 5.976 12.80 FALSE 92
ISOP194BR9P1H4 5.688 394.40 TRUE 93
ISOP194AR9P1D8 5.711 162.07 FALSE 94
ISOP194BR9P1D1 6.696 88.56 TRUE 95
ISOP194AR9P1E8 5.549 570.07 TRUE 96
ISOP194AR9P1E9 5.79  493.72 TRUE 97
ISOP194AR9P1H9 5.694 511.99 TRUE 98
ISOP194BR9P1A9 5.662 225.76 FALSE 99
ISOP194BR9P1A5 7.82  15.28 FALSE 100
ISOP194BR9P1F7 5.982 94.57 TRUE 101
ISOP194AR9P1G7 5.845 50.19 TRUE 102
ISOP194AR9P1E3 6.939 15.65 FALSE 103
ISOP194AR9P1C5 No peak FALSE 104
ISOP194AR9P1H3 6.238 155.66 TRUE 105
ISOP194GR9P1E9 6.343 20.59 TRUE 106
ISOP194HR9P1B10 5.911 398.72 TRUE 107
ISOP194ER9P1A11 5.957 154.65 TRUE 108
ISOP194ER9P1A3 5.976 341.20 TRUE 109
ISOP194ER9P1H9 No peak FALSE 110
ISOP194HR9P1B2 6.274 2.33 FALSE 111
ISOP194HR9P1D11 6.002 433.98 FALSE 112
ISOP194GR9P1F6 6.12  29.42 TRUE 113
ISOP194GR9P1F9 No peak FALSE 114
ISOP194GR9P1C11 12.458  2.90 FALSE 115
ISOP194ER9P1E6 6.125 149.28 TRUE 116
ISOP194BR9P1G9 6.622 84.28 FALSE 117
ISOP194BR9P1E4 5.714 456.33 TRUE 118
ISOP194AR9P1H1 6.247 12.76 FALSE 119
ISOP194BR9P1D10 6.059 10.60 FALSE 120
ISOP194BR9P1C8 No peak FALSE 121
ISOP194AR9P1C10 5.715 98.64 TRUE 122
ISOP194AR9P1D11 No peak FALSE 123
ISOP194AR9P1C3 5.588 700.26 TRUE 124
Sequences
SEQ ID No. 1 = Original Tencon Sequence
LPAPKNLVVSEVTEDSLRLSWTAPDAAFDSFLIQYQESEKVGEAINLTVPGSERSY
DLTGLKPGTEYTVSIYGVKGGHRSNPLSAEFTT
SEQ ID No. 2 = TCL1 library
LPAPKNLVVSEVTEDSLRLSWTAPDAAFDSFLIQYQESEKVGEAINLTVPGSERSY
DLTGLKPGTEYTVSIYGVX1X2X3X4X5X6X7X8X9X10X11X12 PLSAEFTT;
wherein
X1, X2, X3, X4, X5, X6, X7 is any amino acid; and
X8, X9, X10, X11 and X12 are any amino acid or deleted
SEQ ID No. 3 = TCL2 library
LPAPKNLVVSEVTEDSLRLSWX1X2X3X4X5X6X7X8SFLIQYQESEKVGEAINLTVPGS
ERSYDLTGLKPGTEYTVSIYGVX9X10X11X12X135X14X15LSAEFTT;
wherein
X1 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or
Val;
X2 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or
Val;
X3 Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or
Val;
X4 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or
Val;
X5 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or
Val;
X6 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or
Val;
X7 is Phe, Ile, Leu, Val or Tyr;
X8 is Asp, Glu or Thr;
X9 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or
Val;
X10 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or
Val;
X11 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or
Val;
X12 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or
Val;
X13 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or
Val;
X14 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or
Val; and
X15 is Ala, Arg, Asn, Asp, Glu, Gln, Gly, His, Ile, Leu, Lys, Phe, Pro, Ser, Thr, Trp, Tyr or
Val.
SEQ ID No. 4 = Stabilized Tencon
LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFLIQYQESEKVGEAIVLTVPGSERSY
DLTGLKPGTEYTVSIYGVKGGHRSNPLSAIFTT
SEQ ID No. 5 = TCL7 (FG and BC loops)
LPAPKNLVVSRVTEDSARLSWX1X2X3X4X5X6X7X8X9FDSFLIQYQESEKVGEAIVLT
VPGSERSYDLTGLKPGTEYTVSIYGVX10X11X12X13X14X15X16X17X18X19SNPLSAIFTT;
wherein
X1, X2, X3, X4, X5, X6, X10, X11, X12, X13, X14, X15 and X16 are A, D, E, F, G, H, I, K, L, N, P,
Q, R, S, T, V, W or Y;
and
X7, X8, X9, X17, X18 and X19, are A, D, E, F, G, H, I, K, L, N, P, Q, R, S, T, V, W, Y or
deleted
SEQ ID No. 6 = TCL9 (FG loop)
LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFLIQYQESEKVGEAIVLTVPGSERSY
DLTGLKPGTEYTVSIYGVX1X2X3X4X5X6X7X8X9X10X11X12SNPLSAIFTT;
wherein
X1, X2, X3, X4, X5, X6 and X7, is A, D, E, F, G, H, I, K, L, N, P, Q, R, S, T, V, W or Y;
and
X8, X9, X10, X11 and X12 is A, D, E, F, G, H, I, K, L, N, P, Q, R, S, T, V, W, Y or deleted.
TCL14 library (SEQ ID NO: 7):
LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFX1IX2YX3EX4X5X6X7GEAIVLTVPGS
ERSYDLTGLKPGTEYX8VX9IX10GVKGGX11X12SX13PLSAIFTT;
wherein
X1, X2, X3, X4, X5, X6, X7, X8, X9, X10, X11, X12 and X13 are A, D, E, F, G, H, I, K, L, N, P,
Q, R, S, T, V, W, Y, C or M.
TCL24 Library (SEQ ID NO: 8)
LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFX1IX2YX3EX4X5X6X7GEAIX8LX9VPG
SERSYDLTGLKPGTEYX10VX11IX12GVKGGX13X14SX15PLX16AX17FTT;
wherein
X1, X2, X3, X4, X5, X6, X10, X11, X12, X13, X14, X15, X16 and X17 are A, D, E, F, G, H, I, K, L,
N, P, Q, R, S, T, V, Y or W.
SEQ ID No. 9 = Sloning-FOR
GTGACACGGCGGTTAGAAC
SEQ ID No. 10 =  Sloning-REV
GCCTTTGGGAAGCTTCTAAG
SEQ ID No. 11 = POP2250
CGGCGGTTAGAACGCGGCTACAATTAATAC
SEQ ID No. 12 = DigLigRev
CATGATTACGCCAAGCTCAGAA
SEQ ID No. 13 =  BC9
GTGACACGGCGGTTAGAACGCGGCTACAATTAATACATAACCCCATCCCCCTG
TTGACAATTAATCATCGGCTCGTATAATGTGTGGAATTGTGAGCGGATAACAA
TTTCACACAGGAAACAGGATCTACCATGCTGCCGGCGCCGAAAAACCTGGTTG
TTTCTGAAGTTACCGAAGACTCTCTGCGTCTGTCTTGGNNNNNNNNNNNNNNN
NNNNNNNNNNNNTTYGACTCTTTCCTGATCCAGTACCAGGAATCTGAAAAAGT
TGGTGAAGCGATCAACCTGACCGTTCCGGGTTCTGAACGTTCTTACGACCTGA
CCGGTCTGAAACCGGGTACCGAATACACCGTTTCTATCTACGGTGTTCTTAGA
AGCTTCCCAAAGGC
SEQ ID No. 14 = BC8
GTGACACGGCGGTTAGAACGCGGCTACAATTAATACATAACCCCATCCCCCTG
TTGACAATTAATCATCGGCTCGTATAATGTGTGGAATTGTGAGCGGATAACAA
TTTCACACAGGAAACAGGATCTACCATGCTGCCGGCGCCGAAAAACCTGGTTG
TTTCTGAAGTTACCGAAGACTCTCTGCGTCTGTCTTGGNNNNNNNNNNNNNNN
NNNNNNNNNTTYGACTCTTTCCTGATCCAGTACCAGGAATCTGAAAAAGTTGG
TGAAGCGATCAACCTGACCGTTCCGGGTTCTGAACGTTCTTACGACCTGACCG
GTCTGAAACCGGGTACCGAATACACCGTTTCTATCTACGGTGTTCTTAGAAGC
TTCCCAAAGGC
SEQ ID No. 15 = BC7
GTGACACGGCGGTTAGAACGCGGCTACAATTAATACATAACCCCATCCCCCTG
TTGACAATTAATCATCGGCTCGTATAATGTGTGGAATTGTGAGCGGATAACAA
TTTCACACAGGAAACAGGATCTACCATGCTGCCGGCGCCGAAAAACCTGGTTG
TTTCTGAAGTTACCGAAGACTCTCTGCGTCTGTCTTGGNNNNNNNNNNNNNNN
NNNNNNTTYGACTCTTTCCTGATCCAGTACCAGGAATCTGAAAAAGTTGGTGA
AGCGATCAACCTGACCGTTCCGGGTTCTGAACGTTCTTACGACCTGACCGGTC
TGAAACCGGGTACCGAATACACCGTTTCTATCTACGGTGTTCTTAGAAGCTTCC
CAAAGGC
SEQ ID No. 16 = BC6
GTGACACGGCGGTTAGAACGCGGCTACAATTAATACATAACCCCATCCCCCTG
TTGACAATTAATCATCGGCTCGTATAATGTGTGGAATTGTGAGCGGATAACAA
TTTCACACAGGAAACAGGATCTACCATGCTGCCGGCGCCGAAAAACCTGGTTG
TTTCTGAAGTTACCGAAGACTCTCTGCGTCTGTCTTGGNNNNNNNNNNNNNNN
NNNTTYGACTCTTTCCTGATCCAGTACCAGGAATCTGAAAAAGTTGGTGAAGC
GATCAACCTGACCGTTCCGGGTTCTGAACGTTCTTACGACCTGACCGGTCTGA
AACCGGGTACCGAATACACCGTTTCTATCTACGGTGTTCTTAGAAGCTTCCCA
AAGGC
SEQ ID No. 17 = 130 mer-L17A
CGGCGGTTAGAACGCGGCTACAATTAATACATAACCCCATCCCCCTGTTGACA
ATTAATCATCGGCTCGTATAATGTGTGGAATTGTGAGCGGATAACAATTTCAC
ACAGGAAACAGGATCTACCATGCTG
SEQ ID No. 18 = POP222ext
CGG CGG TTA GAA CGC GGC TAC AAT TAA TAC
SEQ ID No. 19 = LS1114
CCA AGA CAG ACG GGC AGA GTC TTC GGT AAC GCG AGA AAC AAC CAG
GTT TTT CGG CGC CGG CAG CAT GGT AGA TCC TGT TTC
SEQ ID No. 20 = LS1115
CCG AAG ACT CTG CCC GTC TGT CTT GG
SEQ ID No. 21 = LS1117
CAG TGG TCT CAC GGA TTC CTG GTA CTG GAT CAG GAA AGA GTC GAA
SEQ ID No. 22 = SDG10
CATGCGGTCTCTTCCGAAAAAGTTGGTGAAGCGATCGTCCTGACCGTTCCGGG
T
SEQ ID No. 23 = SDG24
GGTGGTGAAGATCGCAGACAGCGGGTTAG
SEQ ID No. 24 = POP2222
CGGCGGTTAGAACGCGGCTAC
SEQ ID No. 25 = SDG28
AAGATCAGTTGCGGCCGCTAGACTAGAACCGCTGCCACCGCCGGTGGTGAAG
ATCGCAGAC
SEQ ID No. 26 = FG12
GTGACACGGCGGTTAGAACGCGGCTACAATTAATACATAACCCCATCCCCCTG
TTGACAATTAATCATCGGCTCGTATAATGTGTGGAATTGTGAGCGGATAACAA
TTTCACACAGGAAACAGGATCTACCATGCTGCCGGCGCCGAAAAACCTGGTTG
TTTCTCGCGTTACCGAAGACTCTGCGCGTCTGTCTTGGACCGCGCCGGACGCG
GCGTTCGACTCTTTCCTGATCCAGTACCAGGAATCTGAAAAAGTTGGTGAAGC
GATCGTGCTGACCGTTCCGGGTTCTGAACGTTCTTACGACCTGACCGGTCTGA
AACCGGGTACCGAATACACCGTTTCTATCTACGGTGTTNNNNNNNNNNNNNNN
NNNNNNNNNNNNNNNNNNNNNNNTCTAACCCGCTGTCTGCGATCTTCACCACC
GGCGGTCACCATCACCATCACCATGGCAGCGGTTCTAGTCTAGCGGCCGCAAC
TGATCTTGGC
SEQ ID No. 27 = FG11
GTGACACGGCGGTTAGAACGCGGCTACAATTAATACATAACCCCATCCCCCTG
TTGACAATTAATCATCGGCTCGTATAATGTGTGGAATTGTGAGCGGATAACAA
TTTCACACAGGAAACAGGATCTACCATGCTGCCGGCGCCGAAAAACCTGGTTG
TTTCTCGCGTTACCGAAGACTCTGCGCGTCTGTCTTGGACCGCGCCGGACGCG
GCGTTCGACTCTTTCCTGATCCAGTACCAGGAATCTGAAAAAGTTGGTGAAGC
GATCGTGCTGACCGTTCCGGGTTCTGAACGTTCTTACGACCTGACCGGTCTGA
AACCGGGTACCGAATACACCGTTTCTATCTACGGTGTTNNNNNNNNNNNNNNN
NNNNNNNNNNNNNNNNNNNNTCTAACCCGCTGTCTGCGATCTTCACCACCGGC
GGTCACCATCACCATCACCATGGCAGCGGTTCTAGTCTAGCGGCCGCAACTGA
TCTTGGC
SEQ ID No. 28 = FG10
GTGACACGGCGGTTAGAACGCGGCTACAATTAATACATAACCCCATCCCCCTG
TTGACAATTAATCATCGGCTCGTATAATGTGTGGAATTGTGAGCGGATAACAA
TTTCACACAGGAAACAGGATCTACCATGCTGCCGGCGCCGAAAAACCTGGTTG
TTTCTCGCGTTACCGAAGACTCTGCGCGTCTGTCTTGGACCGCGCCGGACGCG
GCGTTCGACTCTTTCCTGATCCAGTACCAGGAATCTGAAAAAGTTGGTGAAGC
GATCGTGCTGACCGTTCCGGGTTCTGAACGTTCTTACGACCTGACCGGTCTGA
AACCGGGTACCGAATACACCGTTTCTATCTACGGTGTTNNNNNNNNNNNNNNN
NNNNNNNNNNNNNNNNTCTAACCCGCTGTCTGCGATCTTCACCACCGGCGGTC
ACCATCACCATCACCATGGCAGCGGTTCTAGTCTAGCGGCCGCAACTGATCTT
GGC
SEQ ID No. 29 = FG9
GTGACACGGCGGTTAGAACGCGGCTACAATTAATACATAACCCCATCCCCCTG
TTGACAATTAATCATCGGCTCGTATAATGTGTGGAATTGTGAGCGGATAACAA
TTTCACACAGGAAACAGGATCTACCATGCTGCCGGCGCCGAAAAACCTGGTTG
TTTCTCGCGTTACCGAAGACTCTGCGCGTCTGTCTTGGACCGCGCCGGACGCG
GCGTTCGACTCTTTCCTGATCCAGTACCAGGAATCTGAAAAAGTTGGTGAAGC
GATCGTGCTGACCGTTCCGGGTTCTGAACGTTCTTACGACCTGACCGGTCTGA
AACCGGGTACCGAATACACCGTTTCTATCTACGGTGTTNNNNNNNNNNNNNNN
NNNNNNNNNNNNNTCTAACCCGCTGTCTGCGATCTTCACCACCGGCGGTCACC
ATCACCATCACCATGGCAGCGGTTCTAGTCTAGCGGCCGCAACTGATCTTGGC
SEQ ID No. 30 = FG8
GTGACACGGCGGTTAGAACGCGGCTACAATTAATACATAACCCCATCCCCCTG
TTGACAATTAATCATCGGCTCGTATAATGTGTGGAATTGTGAGCGGATAACAA
TTTCACACAGGAAACAGGATCTACCATGCTGCCGGCGCCGAAAAACCTGGTTG
TTTCTCGCGTTACCGAAGACTCTGCGCGTCTGTCTTGGACCGCGCCGGACGCG
GCGTTCGACTCTTTCCTGATCCAGTACCAGGAATCTGAAAAAGTTGGTGAAGC
GATCGTGCTGACCGTTCCGGGTTCTGAACGTTCTTACGACCTGACCGGTCTGA
AACCGGGTACCGAATACACCGTTTCTATCTACGGTGTTNNNNNNNNNNNNNNN
NNNNNNNNNNTCTAACCCGCTGTCTGCGATCTTCACCACCGGCGGTCACCATC
ACCATCACCATGGCAGCGGTTCTAGTCTAGCGGCCGCAACTGATCTTGGC
SEQ ID No. 31 = FG7
GTGACACGGCGGTTAGAACGCGGCTACAATTAATACATAACCCCATCCCCCTG
TTGACAATTAATCATCGGCTCGTATAATGTGTGGAATTGTGAGCGGATAACAA
TTTCACACAGGAAACAGGATCTACCATGCTGCCGGCGCCGAAAAACCTGGTTG
TTTCTCGCGTTACCGAAGACTCTGCGCGTCTGTCTTGGACCGCGCCGGACGCG
GCGTTCGACTCTTTCCTGATCCAGTACCAGGAATCTGAAAAAGTTGGTGAAGC
GATCGTGCTGACCGTTCCGGGTTCTGAACGTTCTTACGACCTGACCGGTCTGA
AACCGGGTACCGAATACACCGTTTCTATCTACGGTGTTNNNNNNNNNNNNNNN
NNNNNNNTCTAACCCGCTGTCTGCGATCTTCACCACCGGCGGTCACCATCACC
ATCACCATGGCAGCGGTTCTAGTCTAGCGGCCGCAACTGATCTTGGC
SEQ ID NO: 32 = human mature PD-L1
FTVTVPKDLYVVEYGSNMTIECKFPVEKQLDLAALIVYWEMEDKNIIQFVHGEED
LKVQHSSYRQRARLLKDQLSLGNAALQITDVKLQDAGVYRCMISYGGADYKRIT
VKVNAPYNKINQRILVVDPVTSEHELTCQAEGYPKAEVIWTSSDHQVLSGKTTTT
NSKREEKLFNVTSTLRINTTTNEIFYCTFRRLDPEENHTAELVIPELPLAHPPNER
SEQ ID NO: 33 = human mature PD-1
PGWFLDSPDRPWNPPTFSPALLVVTEGDNATFTCSFSNTSESFVLNWYRMSPSNQT
DKLAAFPEDRSQPGQDCRFRVTQLPNGRDFHMSVVRARRNDSGTYLCGAISLAPK
AQIKESLRAELRVTERRAEVPTAHPSPSPRPAGQFQTLVVGVVGGLLGSLVLLVW
VLAVICSRAARGTIGARRTGQPLKEDPSAVPVFSVDYGELDFQWREKTPEPPVPCV
PEQTEYATIVFPSGMGTSSPARRGSADGPRSAQPLRPEDGHCSWPL
SEQ
Clone ID NO: AA Sequence
ISOP121HR5P1G9 34 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFPINYGERATKGE
AINLYVPGSERSYDLTGLKPGTEYWVLIGGVKGGLKSSPLWAW
FTT
ISOP121BR5P1F7 35 LPAPKNLVVSRVTEDSARLSWHDATWQYFDSFLIQYQESEKVG
EAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVFHRKHIDFVSNPLS
AIFTT
ISOP121BR5P1A6 36 LPAPKNLVVSRVTEDSARLSWASWLVAFFDSFLIQYQESEKVG
EAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYQRHASAFVSNPL
SAIFTT
ISOP121BR5P1C5 37 LPAPKNLVVSRVTEDSARLSWFRLRIVQTFDSFLIQYQESEKVG
EAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVITVVELLQQSNPLS
AIFTT
ISOP121BR5P1D7 38 LPAPKNLGCFSRYRRLSRLSWETPYPSLSNFDSFLIQYQESEKVG
EAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVLKLSAAWWPSNP
LSAIFTT
ISOP121BR5P1C6 39 LPAPKNLVVSRVTEDSARLSWRKQEQYFDSFLIQYQESEKVGE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYSRPKAEFTSNPLSA
IFTT
ISOP121AR5P1G6 40 LPAPKNLVVSRVTEDSARLSWHATFGDPFDSFLIQYQESEKVG
EAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVGRHYTVYDSNPLS
AIFTT
ISOP121BR5P1B7 41 LPAPKNLVVSRITEDSARLSWKWEEGFFDSFLIQYQESEKVGEA
IVLTVPGSERSYDLTGLKPGTEYTVSIYGVYQRHASAFVSNPLSA
IFTT
ISOP121FR5P1G1 42 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFWIHYTEAPVHGE
AIVLTVPGSERSYDLTGLKPGTEYTVVIWGVKGGTWSSPLSAIF
TT
ISOP121GR5P1B4 43 LPAPKNLIVSRVTEDSARLSWTAPDAAFDSFPINYGERATKGEA
INLYVPGSERSYDLTGLKPGTEYWVLIGGVKGGLKSSPLWAWF
TT
ISOP121BR5P1G2 44 LPAPKNLVVSRVTEDSARLSWADELHHANHFDSFLIQYQESEK
VGEAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYDRHYEIHFYS
NPLSAIFTT
ISOP121HR5P1H2 45 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFDIYYLEYDYSGEA
IVLTVPGSERSYDLTGLKPGTEYDVLIIGVKGGSLSTPLSAIFTT
ISOP121FR5P1G11 46 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFSIWYLEIVAHGE
AIVLTVPGSERSYDLTGLKPGTEYEVIIHGVKGCGPSGPLSAIFTT
ISOP121AR5P1E7 47 LPAPKNLVVSRVTEDSARLSWHVYHEIDYFDSFLIQYQESEKVG
EAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYQRKVEFYSNPLS
AIFTT
ISOP121GR5P1F6 48 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFDIRYHEYTWPGE
AIVLLVPGSERSYDLTGLKPGTEYGVYINGVKGGFRSKPLFAWF
TTGG
ISOP121BR5P1E9 49 LPAPKNLVVSRVTEDSARLSWDSYRDYFDSFLIQYQESEKVGEA
IVLTVPGSERSYDLTGLKPGTEYTVSIYGVYSRKHVVFVQSNPLS
AIFTT
ISOP121AR5P1F2 50 LPAPKNLVISRVTEDSARLSWGWSELIATHFDSFLIQYQESEKV
GEAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYNRKVNFYSNPL
SAIFTT
ISOP121AR5P1F7 51 LPAPKNLVVSRVTEDSARLSWQEHWDTSSNFDSFLIQYQESEK
VGEAIVLTVPGSERSYDLTGLKPGTEYTISIYGVYNRKVLFYSNPL
SAIFTT
ISOP121BR5P1H6 52 LPAPKNLVVSRVTEDSARLSWGYIDVSYFDSFLIQYQESEKVGE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYSRPKAEFTSNPLSA
IFTT
ISOP121GR5P1A2 53 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFKIQYIERYIPGEAI
QLNVPGSERSYDLTGLKPGTEYSVIIPGVKGGRNSFPLWAWFT
T
ISOP121BR5P1D3 54 LPAPKNLVVSRVTEDSARLSWYEDNTERFDSFLIQYQESEKVGE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYIRVQVLWFSNPLS
AIFTT
ISOP121AR5P1F9 55 LPAPKNLVVSRVTEDSARLSWGWSELIATHFDSFLIQYQESEKV
GEAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYNRKVNFYSNPL
SAIFTT
ISOP121AR5P1H5 56 LPAPKNLVVSRVTEDSARLSWEDAVKHIWFDSFLIQYQESEKV
GEAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVWIASVWRSNPL
SAIFTT
ISOP121AR5P1G10 57 LPAPKNLVVSRVTEDSARLSWEWLEHFDSFLIQYQESEKVGEAI
VLTVPGSERSYDLTGLKPGTEYTVSIYGVYQRKVEFHSNPLSAIF
TTT
ISOP121AR5P1F3 58 LPAPKNLVVSRVTEDSARLSWPFNNYSEHFDSFLIQYQESEKVG
EAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYERKTAFYSNPLSA
IFTT
ISOP121BR5P1E2 59 LPAPKNLVVSRVTEDSARLSWWFPLEWFDSFLIQYQESEKVGE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYTREHKSVWASNP
LSAIFTT
ISOP121BR5P1D1 60 LPAPKNLVVSRVTEDSARLSWKWGGEFFDSFLIQYQESEKVGE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYQRNWHHWYSNP
LSAIFTT
ISOP121BR5P1C9 61 LPAPKNLVVSRVTEDSARLSWIWPDKHEFFDSFLIQYQESEKVG
EAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYDRKYANWSSNP
LSAIFTT
ISOP121GR5P1G11 62 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFQINYHEYGQNG
EAIQLIVPGSERSYDLTGLKPGTEYGVWIWGVKGGIRSKPLWA
FFTT
ISOP121BR5P1A7 63 LPAPKNLVVSRVTEDSARLSWTTAFHNEYFDSFLIQYQESEKVG
EAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYSRPKAEFTSNPLS
AIFTT
ISOP121BR5P1C3 64 LPAPKNLVVSRVTEDSARLSWASARDYFDSFLIQYQESEKVGEA
IVLTVPGSERSYDLTGLKPGTEYTVSIYGVLAIAQITHWFSNPLS
AIFTT
ISOP121AR5P1D11 65 LPAPKNLVVSRVTEDSARLSWEWLEHFDSFLIQYQESEKVGEAI
VLTVPGSERSYDLTGLKPGTEYTVSIYGVYQRKVEFHSNPLSAIF
TT
ISOP121ER5P1E7 66 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFTIGYTETPPRGEA
IVLTVPGSERSYDLTGLKPGTKYYVSILGVKGGLGSWPLSAIFTT
ISOP121GR5P1G7 67 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFHIRYHEYDKNGE
AIQLYVPGSERSYDLTGLKPGTEYGVYIHGVKGGGRSKPLWAH
FTT
ISOP121AR5P1A8 68 LPAPKNLVVSRVTEDSARLSWGLEWAYQFFDSFLIQYQESEKV
GEAIVLTVPGSERSYDLTGLKPGTEYIVSIYGVYLRAIEFYSNPLS
AIFTT
ISOP121BR5P1E7 69 LPAPKNLVVSRVTEDSARLSWRKQEQYFDSFLIQYQESEKVGE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVKKWPSTTTTSNPLS
AIFTT
ISOP121FR5P1H8 70 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFVIYYSEQHFYGE
AIVLTVPGSERSYDLTGLKPGTEYVVKIYGVKGGETSKPLSAIFTT
ISOP121GR5P1D2 71 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFHILYQERAQSGE
AIGLVVPGSERSYDLTGLKPATEYSVQIFGVKGGKLSNPLWAW
FTT
ISOP121AR5P1H2 72 LPAPKNLVVSRVTEDSARLSWVIDEFIPLFDSFLIQYQESEKVGE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVLAKNIGISNPLSAIFT
T
ISOP121GR5P1F10 73 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFFIDYVERATVGE
AIALNVPGSKRSYALTGLKPGTEYFVKIRGVKGGLKSKPLWAW
FTT
ISOP121BR5P1A2 74 LPAPKNLVVSRVTEDSARLSWRFSQEWFDSFLIQYQESEKVGE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYARGIHKWLSNPLS
AIFTT
ISOP121GR5P1F7 75 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFGINYVERASEGE
AIDLGVPGSERSYDLTGLKPGTEYFVKIFGVKGGIPSVPLWAWF
TT
ISOP121AR5P1B8 76 LPAPKNLVISRVTEDSARLSWDKRTQFAFDSFLIQYQESEKVGE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVPTWSGRTQSNPLS
AIFTT
ISOP121GR5P1D7 77 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFKIWYQERSIVGE
AIFLLVPGSERSYDLTGLKPGTEYIVQIFGVKGGPYSNPLWAPFT
T
ISOP121BR5P1G3 78 LPAPKNLVVSRVTEDSARLSWKQRTSFHFDSFLIQYQESEKVGE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVPFWQQWQPESNP
LSAIFTT
ISOP121AR5P1C5 79 LPAPKNLVVSRVTEDSARLSWKRSDDEWFDSFLIQYQESEKVG
EAIILTVPGSERSYDLTGLKPGTEYTVSIYGVYQRAALWFSNPLS
AIFTT
ISOP121FR5P1H9 80 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFSILYGETAPIGEAI
VLTVPGSERSYDLTGLKPGTEYVVYIQGVKGGNYSQPLSAIFTT
ISOP121AR5P1A10 81 LPAPKNLVVSRVTEDSARLSWPDWSNSEYFDSFLIQYQESEKV
GEAIVLTVPGSERSYDLTGLKPGTEYIVSIYGVYARHRLFVSNPL
SAIFTT
ISOP121HR5P1F2 82 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFTILYGETYSGGEA
IVLTVPGSERSYDLTGLKPGTEYVVYIFGVKGGKWSRPLSAIFTT
ISOP121AR5P1H1 83 LPAPKNLVVSRVTEDSARLSWKQATKFVFDSFLIQYQESEKVGE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVPDWFVLESNPLSAI
FTT
ISOP121BR5P1D10 84 LPAPKNLVVSRVTEDSARLSWGKKSHFFDSFLIQYQESEKVGEA
IVLTVPGSERSYDLTGLKPGTEYTVSIYGVYTRGQCEWESNQLS
AIFFTT
ISOP121BR5P1F10 85 LPAPKNLVVSRVTEDSARLSWPLNLEYFDSFLIQYQESEKVGEAI
VLTVPGSERSYDLTGLKPGTEYTVSIYGVYGRYGGPFVSNPLSAI
FTT
ISOP121BR5P1D11 86 LPAPKNLVVSRVTEDSARLSWFNADEEYFDSFLIQYQESEKVGE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYVRAVRFVSSNPLS
AIFTT
ISOP121AR5P1E11 87 LPAPKNLVVSRVTEDSARLSWSVQTSFVFDSFLIQYQESEKVGE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVPLWHGFDSNPLSAI
FTT
ISOP121BR5P1D6 88 LPAPKNLVVSRVTEDSARLSWKQGTSFHFDSFLIQYQESEKVGE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVQLLANDIISSNPLSAI
FTT
ISOP121BR5P1B5 89 LPAPKNLVVSRVTEDSARLSWRKQEQYFDSFLIQYQESEKVGE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYQRGYHNWFSNPL
SAIFTT
ISOP194ER9P1G3 90 LPAPKNLIVSRVTEDSARLSWTAPDAAFDSFRIAYYETMVSGEA
IVLTVPGSERSYDLTGLKPGTEYAVIIKGVKGGKPSWPLSAIFTT
ISOP194AR9P1F2 91 LPAPKNLVISRVTEDSARLSWEWLEHFDSFLIQYQESEKVGEAI
VLTVPGSERSYDLTGLKPGTEYTVSIYGVYNRKVNFYSNPLSAIF
TT
ISOP194AR9P1H10 92 LPAPKNLVISRVTEDSARLSWPAHYHSAFFDSFLIQYQESEKVG
EAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYQRKVEFHSNPLS
AIFTT
ISOP194BR9P1H4 93 LPAPKNLVVSRVTEDSACLSWTTAFHNEYFDSFLIQYQESEKVG
EAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYSRPKAEFTSNPLS
AIFTT
ISOP194AR9P1D8 94 LPAPKNLVVSRVTEDSARLSWDTWNDFFDSFLIQYQESEKVGE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYQRKVIWLSNPLSA
IFTT
ISOP194BR9P1D1 95 LPAPKNLVVSRVTEDSARLSWEHSLLNDQWFDSFLIQYQESEK
VGEAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYQRGRALWYS
NPLSAIFTT
ISOP194AR9P1E8 96 LPAPKNLVVSRVTEDSARLSWEWLEHFDSFLIQYQESEKVGEAI
VLTIPGSERSYDLTGLKPGTEYTVSIYGVYQRKVEFHSNPLSAIFT
T
ISOP194AR9P1E9 97 LPAPKNLVVSRVTEDSARLSWEWLEHFDSFLIQYQESEKVGEAI
VLTVPGSERSYDLTGLKPGTEYTVSIYGVYQRKVNFYSNPLSAIF
TT
ISOP194AR9P1H9 98 LPAPKNLVVSRVTEDSARLSWEWLEHFDSFQIQYQESEKVGEA
IVLTVPGSERSYDLTGLKPGTEYIVSIYGVYQRKVEFHSNPLSAIF
TT
ISOP194BR9P1A9 99 LPAPKNLVVSRVTEDSARLSWFNADEEYFDSFLIQYQESEKVGE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYDRKVKFVQSNPLS
AIFTT
ISOP194BR9P1A5 100 LPAPKNLVVSRVTEDSARLSWFNADEEYFDSFLIQYQESEKVGE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYQRGYHNWFSNPL
SAIFTT
ISOP194BR9P1F7 101 LPAPKNLVVSRVTEDSARLSWFNADEEYFDSFLIQYQESEKVGE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYTRGRYEWRESNPL
SAIFTT
ISOP194AR9P1G7 102 LPAPKNLVVSRVTEDSARLSWGDDFNSEYFDSFLIQYQESEKV
GEAIVLTVPGSERSYDLTGLKPGTEYIVSIYGVYTRAVVFTSNPL
SAIFTT
ISOP194AR9P1E3 103 LPAPKNLVVSRVTEDSARLSWKRSDDEWFDSFLIQYQESEKVG
EAIVLTVPGSERSYDLTGLKPGTEYIVSIYGVYQRAALWFSNPLS
AIFTT
ISOP194AR9P1C5 104 LPAPKNLVVSRVTEDSARLSWLRDFNGRAFFDSFLIQYQESEKV
GEAIVLTVPGSERSYDPTGLKPGTEYIVSIYGVFITWIHVRSNPL
SAIFTT
ISOP194AR9P1H3 105 LPAPKNLVVSRVTEDSARLSWNASWISHNFFDSFLIQYQESEK
VGEAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYERKTAFYSNP
LSAIFTT
ISOP194GR9P1E9 106 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFHIRYHEYDKNGE
AIQLYVPGSERSYDLTGLKPGTEYGVFIWGVKGGLKSKPLWAW
FTT
ISOP194HR9P1B10 107 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFPIRYYERANGEAI
VLTVPGSERSYDLTGLKPGTEYIVWIYGVKGGGRSGPLSAIFTT
ISOP194ER9P1A11 108 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFRIAYYETMVSGE
AIVLTVPGSERSYDLTGLKPGTEYAVIIKGVKGGKPSWPLSAIFT
T
ISOP194ER9P1A3 109 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFRIAYYETMVSGE
AIVLTVPGSERSYDLTGLKPGTEYAVIIKGVKGGMVSWPLSAIFT
T
ISOP194ER9P1H9 110 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFRIAYYETMVSGE
AIVLTVPGSERSYDLTGPKPGTEYAVIIKGVKGGKPSWPLSAIFT
T
ISOP194HR9P1B2 111 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFSILYGELIGDGEAI
VLTVPGSERSYDLTGLKPGSEYTVYIFGVKGGRYSRPLSAIFTT
ISOP194HR9P1D11 112 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFSILYGELIGDGEAI
VLTVPGSERSYDLTGLKPGTEYTVYIFGVKGGRYSRPLSAIFTT
ISOP194GR9P1F6 113 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFWIDYWERLSEGE
AIALRVPGSERSYDLTGLKPGTEYYVWIVGVKGGKFSQPLRAW
FTT
ISOP194GR9P1F9 114 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFWIFYNERWQNG
EAIRLIVPGSERSYDLTGLKPGTEYSVIIPGVKGGRNSFPLWAWF
TT
ISOP194GR9P1C11 115 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFWIFYNERWQNG
EAIRLTVPGSERSYDLTGLKPGTEYWVLIGGVKGGLKSSPLWA
WFTT
ISOP194ER9P1E6 116 LPAPKNLVVSRVTEDSARLSWTAPDAAFDSFWIKYYEKRNPGE
AIVLTVPGSERSYDLTGLKPGTEYLVIISGVKGGSRSVPLSAIFTT
ISOP194BR9P1G9 117 LPAPKNLVVSRVTEDSARLSWTTAFHNEYFDSFLIQYQESEKVG
EAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYIRVQVLWFSNPL
SAIFTT
ISOP194BR9P1E4 118 LPAPKNLVVSRVTEDSARLSWTTAFHNEYFDSFLIQYQESEKVG
EAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYQRGYHNWFSNP
LSAIFTT
ISOP194AR9P1H1 119 LPAPKNLVVSRVTEDSARLSWWRVLGHSHFFDSFLIQYQESEK
VGEAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYNRKVNFYSNP
LSAIFTT
ISOP194BR9P1D10 120 LPAPKNLVVSRVTEDSARLSWYEDNTERFDSFLIQYQESEKVVE
AIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYIRVQVLWFSNPLS
AIFTT
ISOP194BR9P1C8 121 LPAPKNLVVSRVTEDSARLSWYFAGELWFDSFLIQYQESEKVG
EAIVLTVPGSERSYDLTGLKPGTEYTVSIYGVYQRGYHNWFSNP
LSAIFTT
ISOP194AR9P1C10 122 LPAPKNLVVSRVTEDSARPSWEWLEHFDSFLIQYQESEKVGEAI
VLTVPGSERSYDLTGLKPGTEYTVSIYGVYNRKVNFYSNPLSAIF
TT
ISOP194AR9P1D11 123 LPAPKNLVVSRVTEDSGRLSWQHHISFFDSFLIQYQESEKVGEA
IVLTVPGSERSYDLTGLKPGTEYTVSIYGVYNRKVNFYSNPLSAIF
TT
ISOP194AR9P1C3 124 LPAPKNLVVSRVTQNSARLSWEWLEHFDSFLIHYQESEKVGEA
IVLTVPGSERSYDLTGLKPGTEYTVSIYGVYQRKVEFHSNPLSAIF
TT
3rd FN3 domain of 125 DAPSQIEVKDVTDTTALITWFKPLAEIDGIELTYGIKDVP
tenascin C (TN3 GDRTTIDLTEDENQYSIGNLKPDTEYEVSLISRR
GDMSSNPAKETFTT
Fibcon 126 LDAPTDLQVTNVTDTSITVSWTPPSATITGYRITYTPSNG
PGEPKELTVPPSSTSVTITGLTPGVEYVVSLYAL
KDNQESPPLVGTQTT
10th FN3 domain of 127 VSDVPRDLEVVAATPTSLLISWDAPAVTVRYYRITYGET
fibronectin GGNSPVQEFTVPGSKSTATISGLKPGVDYTITVY
AVTGRGDSPASSKPISINYRT
Linker 128 GSGS
Linker 129 GGGSGGGS
Linker 130 GGGGSGGGGSGGGGSGGGGSGGGGS
Linker 131 APAP
Linker 132 APAPAPAPAP
Linker 133 APAPAPAPAPAPAPAPAPAP
Linker 134 APAPAPAPAPAPAPAPAPAPAPAPAPAPAPAPAPAPAPAP
Linker 135 EAAAKEAAAKEAAAKEAAAKEAAAKAAA
Albumin variant 136 DAHKSEVAHRFKDLGEENFKALVLIAFAQYLQQSPFED
HVKLVNEVTEFAKTCVADESAENCDKSLHTLFG
DKLCTVATLRETYGEMADCCAKQEPERNECFL
QHKDDNPNLPRLVRPEVDVMCTAFFIDNEETFL
KKYLYEIARRHPYFYAPELLFFAKRYKAAFTEC
CQAADKAACLLPKLDELRDEGKASSAKQRLKC
ASLQKFGERAFKAWAVARLSQRFPKAEFAEVSK
LVTDLTKVHTECCHGDLLECADDRADLAKYICE
NQDSISSKLKECCEKPLLEKSHCIAEVENDEMPA
DLPSLAADFVESKDVCKNYAEAKDVFLGMFLY
EYARRHPDYSVVLLLRLAKTYETTLEKCCAAAD
PHECYAKVFDEFKPLVEEPQNLIKQNCELFEQLG
EYKFQNALLVRYTKKVPQVSTPTLVEVSRNLGK
VGSKCCKHPEAKRMPCAEDYLSVVLNQLCVLH
EKTPVSDRVTKCCTESLVNRRPCFSALEVDETY
VPKEFNAETFTFHADICTLSEKERQIKKQTALVE
LVKHKPKATKEQLKAVMDDFAAFVEKCCKAD
DKETCFAEEGKKLVAASQAALGL

Claims (13)

What is claimed:
1. A protein comprising an amino acid sequence that is at least 95% identical to a sequence selected from the group consisting of SEQ ID NOs: 34, 35, 36, 37, 38, 39, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, and 124.
2. The protein of claim 1, wherein the protein has at least one substitution at a residue selected from the group consisting of 11, 14, 17, 37, 46, 73, and 86 corresponding to SEQ ID NO: 4.
3. The protein of claim 1, wherein the protein is conjugated to a heterologous molecule.
4. The protein of claim 3, wherein the heterologous molecule is a detectable label, a cytotoxic agent, or both.
5. The protein of claim 4, wherein the detectable label is selected from a radioactive isotope, a magnetic bead, a metallic bead, a colloidal particle, a fluorescent dye, an electron-dense reagent, an enzyme, a biotin, a digoxigenin, a hapten, a luminescent molecule, a chemiluminescent molecule, a fluorochrome, a fluorophore, a fluorescent quenching agent, a colored molecule, a scintillant, an avidin, a streptavidin, a protein A, a protein G, an antibody, an antibody fragment, a polyhistidine, a Ni2+, a flag tag, a myc tag, a heavy metal, an alkaline phosphatase, a peroxidase, a luciferase, an electron donor, an electron acceptor, an acridinium ester, or a colorimetric substrate.
6. The protein of claim 4, wherein the detectable label is auristatin, monomethyl auristatin phenylalanine, dolostatin, chemotherapeutic agent, a drug, a growth inhibitory agent, a toxin, or a radioactive isotope.
7. The protein of claim 4, wherein the detectable label is conjugated to the protein by a linker.
8. The protein of claim 4, wherein the detectable label is complexed with a chelating agent.
9. The protein of claim 1, further comprising a methionine at the N-terminus of the protein.
10. The protein of claim 1, wherein the protein is coupled to a half-life extending moiety.
11. The protein of claim 10, wherein the half-life extending moiety is an albumin binding molecule, a polyethylene glycol (PEG), albumin, albumin variant, or at least a portion of an Fc region of an immunoglobulin.
12. A composition comprising the protein of claim 1 and a pharmaceutically acceptable carrier.
13. A kit comprising the protein of claim 1.
US16/801,787 2016-12-14 2020-02-26 PD-L1 binding fibronectin type III domains Active US11447539B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/801,787 US11447539B2 (en) 2016-12-14 2020-02-26 PD-L1 binding fibronectin type III domains

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201662434054P 2016-12-14 2016-12-14
US15/840,281 US10597438B2 (en) 2016-12-14 2017-12-13 PD-L1 binding fibronectin type III domains
US16/801,787 US11447539B2 (en) 2016-12-14 2020-02-26 PD-L1 binding fibronectin type III domains

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/840,281 Continuation US10597438B2 (en) 2016-12-14 2017-12-13 PD-L1 binding fibronectin type III domains

Publications (2)

Publication Number Publication Date
US20210024612A1 US20210024612A1 (en) 2021-01-28
US11447539B2 true US11447539B2 (en) 2022-09-20

Family

ID=62488652

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/840,281 Active US10597438B2 (en) 2016-12-14 2017-12-13 PD-L1 binding fibronectin type III domains
US16/801,787 Active US11447539B2 (en) 2016-12-14 2020-02-26 PD-L1 binding fibronectin type III domains

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US15/840,281 Active US10597438B2 (en) 2016-12-14 2017-12-13 PD-L1 binding fibronectin type III domains

Country Status (3)

Country Link
US (2) US10597438B2 (en)
EP (1) EP3554535A4 (en)
WO (1) WO2018111976A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3554562A4 (en) 2016-12-14 2020-11-04 Janssen Biotech, Inc. Cd8a-binding fibronectin type iii domains
WO2018111976A1 (en) * 2016-12-14 2018-06-21 Janssen Biotech, Inc. Pd-l1 binding fibronectin type iii domains
WO2018111978A1 (en) 2016-12-14 2018-06-21 Janssen Biotech, Inc. Cd137 binding fibronectin type iii domains
WO2021076546A1 (en) 2019-10-14 2021-04-22 Aro Biotherapeutics Company Cd71 binding fibronectin type iii domains
US11781138B2 (en) 2019-10-14 2023-10-10 Aro Biotherapeutics Company FN3 domain-siRNA conjugates and uses thereof

Citations (139)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4281061A (en) 1979-07-27 1981-07-28 Syva Company Double antibody for enhanced sensitivity in immunoassay
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
WO1996038557A1 (en) 1995-06-02 1996-12-05 Genentech, Inc. Hepatocyte growth factor receptor antagonists and uses thereof
US5643768A (en) 1989-10-05 1997-07-01 Optein, Inc. Cell-free synthesis and isolation of novel genes and polypeptides
US5643763A (en) 1994-11-04 1997-07-01 Genpharm International, Inc. Method for making recombinant yeast artificial chromosomes by minimizing diploid doubling during mating
US5658727A (en) 1991-04-10 1997-08-19 The Scripps Research Institute Heterodimeric receptor libraries using phagemids
US5691157A (en) 1995-10-24 1997-11-25 The Research Foundation Of State University Of New York Method for detecting a mammal's prior exposure to radiation or radiomimetic chemicals
US5846456A (en) 1996-01-17 1998-12-08 National Science Council Method of making gradient index optical element
US5856456A (en) 1992-11-20 1999-01-05 Enzon, Inc. Linker for linked fusion polypeptides
US6018030A (en) 1986-11-04 2000-01-25 Protein Polymer Technologies, Inc. Peptides comprising repetitive units of amino acids and DNA sequences encoding the same
EP0985039A2 (en) 1997-06-12 2000-03-15 Research Corporation Technologies, Inc Artificial antibody polypeptides
US6162903A (en) 1992-05-07 2000-12-19 Actinova Limited Immunoglobulin binding proteins derived from L protein and their uses
US6172197B1 (en) 1991-07-10 2001-01-09 Medical Research Council Methods for producing members of specific binding pairs
WO2001014557A1 (en) 1999-08-23 2001-03-01 Dana-Farber Cancer Institute, Inc. Pd-1, a receptor for b7-4, and uses therefor
WO2001064942A1 (en) 2000-02-29 2001-09-07 Phylos, Inc. Protein scaffolds for antibody mimics and other binding proteins
EP1137941A1 (en) 1998-12-10 2001-10-04 Phylos, Inc. Protein scaffolds for antibody mimics and other binding proteins
WO2002032925A2 (en) 2000-10-16 2002-04-25 Phylos, Inc. Protein scaffolds for antibody mimics and other binding proteins
US6472147B1 (en) 1999-05-25 2002-10-29 The Scripps Research Institute Methods for display of heterodimeric proteins on filamentous phage using pVII and pIX, compositions, vectors and combinatorial libraries
US6521427B1 (en) 1997-09-16 2003-02-18 Egea Biosciences, Inc. Method for the complete chemical synthesis and assembly of genes and genomes
US6582915B1 (en) 1991-12-02 2003-06-24 Medical Research Council Production of anti-self bodies from antibody segment repertories and displayed on phage
WO2003104418A2 (en) 2002-06-06 2003-12-18 Research Corporation Technologies, Inc. Reconstituted polypeptides
US6670127B2 (en) 1997-09-16 2003-12-30 Egea Biosciences, Inc. Method for assembly of a polynucleotide encoding a target polypeptide
WO2004029224A2 (en) 2002-09-30 2004-04-08 Compound Therapeutics, Inc. Methods of engineering spatially conserved motifs in polypeptides
WO2004058821A2 (en) 2002-12-27 2004-07-15 Domantis Limited Dual specific single domain antibodies specific for a ligand and for the receptor of the ligand
US20040197332A1 (en) 2001-08-09 2004-10-07 Axel Ullrich Inhibitors of her3 activity
US20040259781A1 (en) 2001-08-09 2004-12-23 Ruth Chiquet-Ehrismann Anti-tumour agents and methods of identifying anti-tumour agents
US20050004029A1 (en) 2001-11-01 2005-01-06 Garcia Joe G.N. Methods and compositions for treating vascular leak using hepatocyte growth factor
US6846655B1 (en) 1998-06-29 2005-01-25 Phylos, Inc. Methods for generating highly diverse libraries
US20050038229A1 (en) 1998-12-10 2005-02-17 Dasa Lipovsek Protein scaffolds for antibody mimics and other binding proteins
WO2005018534A2 (en) 2003-05-16 2005-03-03 Rosetta Inpharmatics, Llc Methods and compositions for rna interference
WO2005042708A2 (en) 2003-10-27 2005-05-12 Rosetta Inpharmatics Llc METHOD OF DESIGNING siRNAS FOR GENE SILENCING
US6969108B2 (en) 2001-09-26 2005-11-29 Aisin Seiki Kabushiki Kaisha Vehicular door
US20050272083A1 (en) 2004-06-04 2005-12-08 Somasekar Seshagiri EGFR mutations
US20060040278A1 (en) 2004-01-27 2006-02-23 Cojocaru Gad S Novel nucleotide and amino acid sequences, and assays and methods of use thereof for diagnosis of ovarian cancer
WO2007000671A2 (en) 2005-06-15 2007-01-04 Monoclonal Antibodies Therapeutics Anti-cd71 monoclonal antibodies and uses thereof for treating malignant tumour cells
US20070148126A1 (en) 2003-12-05 2007-06-28 Yan Chen Inhibitors of type 2 vascular endothelial growth factor receptors
WO2007085815A2 (en) 2006-01-24 2007-08-02 Domantis Limited Ligands that bind il-4 and/or il-13
US20070184476A1 (en) 2006-01-24 2007-08-09 Hui-Chu Hsieh Biomarkers for liver fibrotic injury
US7288638B2 (en) 2003-10-10 2007-10-30 Bristol-Myers Squibb Company Fully human antibodies against human 4-1BB
WO2008079973A2 (en) 2006-12-21 2008-07-03 Centocor, Inc. Egfr binding peptides and uses thereof
US20080220049A1 (en) 2003-12-05 2008-09-11 Adnexus, A Bristol-Myers Squibb R&D Company Compositions and methods for intraocular delivery of fibronectin scaffold domain proteins
US20080241159A1 (en) 2003-04-16 2008-10-02 Genentech, Inc. Methods and compositions for selective modulation of vascularization
WO2008127710A2 (en) 2007-04-13 2008-10-23 Dana Farber Cancer Institute Methods for treating cancer resistant to erbb therapeutics
WO2008156642A1 (en) 2007-06-15 2008-12-24 Vasgene Therapeutics, Inc. Non-immunoglobulin antigen binding scaffolds for inhibiting angiogenesis and tumor growth
US20090042906A1 (en) 2007-04-26 2009-02-12 Massachusetts Institute Of Technology Methods for treating cancers associated with constitutive egfr signaling
WO2009023184A2 (en) 2007-08-10 2009-02-19 Protelix, Inc. Universal fibronectin type iii binding-domain libraries
WO2009058379A2 (en) 2007-10-31 2009-05-07 Medimmune, Llc Protein scaffolds
WO2009086116A2 (en) 2007-12-19 2009-07-09 Centocor, Inc. Alternative scaffold protein fusions phage display via fusion to plx of m13 phage
WO2009085462A1 (en) 2007-12-19 2009-07-09 Centocor, Inc. Design and generation of human de novo pix phage display libraries via fusion to pix or pvii, vectors, antibodies and methods
WO2009083804A2 (en) 2007-12-27 2009-07-09 Novartis Ag Improved fibronectin-based binding molecules and their use
WO2009102421A2 (en) 2008-02-14 2009-08-20 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins that bind egfr
WO2009111691A2 (en) 2008-03-06 2009-09-11 Genentech, Inc. Combination therapy with c-met and egfr antagonists
WO2009126834A2 (en) 2008-04-11 2009-10-15 Galaxy Biotech, Llc Combination of hgf inhibitor and egf inhibitor to treat cancer
WO2009133208A1 (en) 2008-05-02 2009-11-05 Novartis Ag Improved fibronectin-based binding molecules and uses thereof
US20090274693A1 (en) 2008-05-05 2009-11-05 Gilmer Tona M Method of Treating Cancer using a cMet and AXL Inhibitor and an ErbB Inhibitor
WO2009142773A2 (en) 2008-05-22 2009-11-26 Bristol-Myers Squibb Company Multivalent fibronectin based scaffold domain proteins
US20090311803A1 (en) 2006-03-31 2009-12-17 Way Jeffrey C Treatment Of Tumors Expressing Mutant EGF Receptors
WO2010039248A1 (en) 2008-10-01 2010-04-08 Ludwig Institute For Cancer Research Methods for the treatment of cancer
US7709214B2 (en) 2000-06-28 2010-05-04 Dana-Farber Cancer Institute, Inc. Methods for upregulating an immune response with agents that inhibit the intereaction between PD-L2 and PD-1
WO2010051310A2 (en) 2008-10-31 2010-05-06 Centocor Ortho Biotech Inc. ANTI-TNFα FIBRONECTIN TYPE III DOMAIN BASED SCAFFOLD COMPOSITIONS, METHODS AND USES
WO2010051274A2 (en) 2008-10-31 2010-05-06 Centocor Ortho Biotech Inc. Fibronectin type iii domain based scaffold compositions, methods and uses
WO2010060095A1 (en) 2008-11-24 2010-05-27 Bristol-Myers Squibb Company Bispecific egfr/igfir binding molecules
WO2010093627A2 (en) 2009-02-12 2010-08-19 Centocor Ortho Biotech Inc. Fibronectin type iii domain based scaffold compositions, methods and uses
US7794710B2 (en) 2001-04-20 2010-09-14 Mayo Foundation For Medical Education And Research Methods of enhancing T cell responsiveness
WO2010115202A2 (en) 2009-04-03 2010-10-07 Dicerna Pharmaceuticals, Inc. Methods and compositions for the specific inhibition of kras by blunt ended double-stranded rna
US20100254989A1 (en) 2009-04-07 2010-10-07 Birgit Bossenmaier Bispecific Anti ErbB1 / Anti c Met Antibodies
US7842476B2 (en) 2002-09-06 2010-11-30 Isogenica Limited In vitro peptide expression library
WO2011005133A1 (en) 2009-07-07 2011-01-13 Siemens Aktiengesellschaft Apparatus and method for measuring multi-phase fluid flow
US20110081345A1 (en) 2007-04-18 2011-04-07 Moore Margaret D Single chain fc, methods of making and methods of treatment
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
US20110118144A1 (en) 2007-12-19 2011-05-19 Linus Hyun Engineered phage vectors for the design and the generation of a human non-antibody peptide or protein phage library via fusion to pix of m13 phage
WO2011110642A2 (en) 2010-03-10 2011-09-15 Genmab A/S Monoclonal antibodies against c-met
WO2011130324A1 (en) 2010-04-13 2011-10-20 Medimmune, Llc Fibronectin type iii domain-based multimeric scaffolds
WO2011131746A2 (en) 2010-04-20 2011-10-27 Genmab A/S Heterodimeric antibody fc-containing proteins and methods for production thereof
WO2011137319A2 (en) 2010-04-30 2011-11-03 Centocor Ortho Biotech Inc. Stabilized fibronectin domain compositions, methods and uses
US20110287009A1 (en) 2010-04-23 2011-11-24 Genentech, Inc. Production of Heteromultimeric Proteins
WO2011151412A1 (en) 2010-06-01 2011-12-08 Pierre Fabre Medicament Novel anti-cmet antibody
WO2012016245A2 (en) 2010-07-30 2012-02-02 Novartis Ag Fibronectin cradle molecules and libraries thereof
US8217149B2 (en) 2008-12-09 2012-07-10 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
US20120244164A1 (en) 2010-09-30 2012-09-27 Ablynx N.V. Biological materials related to c-met
US20120263723A1 (en) 2008-11-21 2012-10-18 Eli Lilly And Company c-Met Antibodies
US20120270797A1 (en) * 2009-08-13 2012-10-25 Massachusetts Institute Of Technology Engineered proteins including mutant fibronectin domains
WO2012162418A1 (en) 2011-05-23 2012-11-29 The University Of North Carolina At Chapel Hill Methods and compositions for heterodimeric targeting ligands
US20120315639A1 (en) 2011-06-08 2012-12-13 Glenn Yaguang Deng Method and apparatus for single cell isolation and analysis
US20120321666A1 (en) 2011-05-23 2012-12-20 Cooper Laurence J N T cell therapy for b cell lymphoma
US20130039927A1 (en) 2010-02-12 2013-02-14 University Of Rochester Antigenic mimics of discontinuous epitopes of pathogen recognized by broadly neutralizing antibodies
WO2013049275A1 (en) 2011-09-27 2013-04-04 Janssen Biotech, Inc. Fibronectin type iii repeat based protein scaffolds with alternative binding surfaces
US20130184212A1 (en) 2010-05-26 2013-07-18 Bristol-Myers Squibb Company Fibronectin based scaffold proteins having improved stability
US20130226834A1 (en) 2000-04-27 2013-08-29 Networth Services, Inc. Systems and methods for determining the financial status of bonds
US8552154B2 (en) 2008-09-26 2013-10-08 Emory University Anti-PD-L1 antibodies and uses therefor
US20130273561A1 (en) 2010-10-29 2013-10-17 The Governing Council Of The University Of Toronto Lipid encapsulation of surface enhanced raman scattering (sers) nanoparticles
US20140141000A1 (en) 2012-11-21 2014-05-22 Janssen Biotech, Inc. Bispecific EGFR/C-Met Antibodies
WO2014081944A2 (en) 2012-11-21 2014-05-30 Janssen Biotech, Inc. Egfr and c-met-fibronectin type iii domain binding molecules
US8741295B2 (en) 2009-02-09 2014-06-03 Universite De La Mediterranee PD-1 antibodies and PD-L1 antibodies and uses thereof
WO2014100079A1 (en) 2012-12-21 2014-06-26 Merck Sharp & Dohme Corp. Antibodies that bind to human programmed death ligand 1 (pd-l1)
US8779108B2 (en) 2009-11-24 2014-07-15 Medimmune, Limited Targeted binding agents against B7-H1
US20140271467A1 (en) * 2013-03-12 2014-09-18 The Board Of Trustees Of The Leland Stanford Junior University Probes and methods of imaging non-hodgkins lymphoma
WO2014165093A2 (en) 2013-03-13 2014-10-09 Bristol-Myers Squibb Company Fibronectin based scaffold domains linked to serum albumin or a moiety binding thereto
WO2014165082A2 (en) 2013-03-13 2014-10-09 Medimmune, Llc Antibodies and methods of detection
US20140341917A1 (en) 2011-11-28 2014-11-20 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof
WO2014189973A2 (en) 2013-05-20 2014-11-27 Genentech, Inc. Anti-transferrin receptor antibodies and methods of use
US20140371296A1 (en) 2011-12-22 2014-12-18 Isis Pharmaceuticals, Inc. Methods for modulating metastasis-associated-in-lung-adenocarcinoma-transcript-1 (malat-1) expression
WO2014209804A1 (en) 2013-06-24 2014-12-31 Biomed Valley Discoveries, Inc. Bispecific antibodies
US20150005364A1 (en) 2012-01-05 2015-01-01 Bioneer Corporation High-efficiency nanoparticle-type double-helical oligo-rna structure and method for preparing same
US8981063B2 (en) 1999-11-30 2015-03-17 Mayo Foundation For Medical Education And Research B7-H1 Antibodies
US20150104808A1 (en) 2013-10-14 2015-04-16 Janssen Biotech, Inc. Cysteine Engineered Fibronectin Type III Domain Binding Molecules
WO2015061668A1 (en) 2013-10-25 2015-04-30 Dana-Farber Cancer Institute, Inc. Anti-pd-l1 monoclonal antibodies and fragments thereof
US20150118288A1 (en) 2012-05-23 2015-04-30 Ohio State Innovation Foundation Lipid Nanoparticle Compositions and Methods of Making and Methods of Using the Same
WO2015092393A2 (en) 2013-12-17 2015-06-25 Kymab Limited Human targets
US20150197571A1 (en) 2012-08-03 2015-07-16 Dana-Farber Cancer Institute, Inc. Single Agent Anti-PD-L1 and PD-L2 Dual Binding Antibodies and Methods of Use
WO2015109124A2 (en) 2014-01-15 2015-07-23 Kadmon Corporation, Llc Immunomodulatory agents
US20150203580A1 (en) 2014-01-23 2015-07-23 Regeneron Pharmaceuticals, Inc. Human Antibodies to PD-L1
US20150252097A1 (en) 2006-11-22 2015-09-10 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins for tyrosine kinases receptors, including igf-ir
US20150274835A1 (en) 2012-10-04 2015-10-01 Dana-Farber Cancer Institute, Inc. Human monoclonal anti-pd-l1 antibodies and methods of use
US9175082B2 (en) 2012-05-31 2015-11-03 Sorrento Therapeutics, Inc. Antigen binding proteins that bind PD-L1
US20150346208A1 (en) 2014-05-29 2015-12-03 Spring Bioscience Corporation PD-L1 Antibodies and Uses Thereof
US9212224B2 (en) 2012-05-15 2015-12-15 Bristol-Myers Squibb Company Antibodies that bind PD-L1 and uses thereof
WO2015195163A1 (en) 2014-06-20 2015-12-23 R-Pharm Overseas, Inc. Pd-l1 antagonist fully human antibody
WO2016000619A1 (en) 2014-07-03 2016-01-07 Beigene, Ltd. Anti-pd-l1 antibodies and their use as therapeutics and diagnostics
WO2016004043A1 (en) 2014-06-30 2016-01-07 Blend Therapeutics, Inc. Targeted conjugates and particles and formulations thereof
WO2016086036A2 (en) 2014-11-25 2016-06-02 Bristol-Myers Squibb Company Methods and compositions for 18f-radiolabeling of biologics
WO2016086021A1 (en) 2014-11-25 2016-06-02 Bristol-Myers Squibb Company Novel pd-l1 binding polypeptides for imaging
CN105907719A (en) 2016-04-18 2016-08-31 李华顺 Anti BOBO1 CAR-T cell and preparation and application thereof
US20160303256A1 (en) 2013-09-30 2016-10-20 The University Of North Carolina At Chapel Hill Methods and compositions for self-assembly system of nanoparticles and microparticles for multi-targeting specificity
US20160326232A1 (en) 2015-05-06 2016-11-10 Janssen Biotech, Inc. Prostate Specific Membrane Antigen Binding Fibronectin Type III Domains
WO2016197071A1 (en) 2015-06-05 2016-12-08 New York University Compositions and methods for anti-staphylococcal biologic agents
US20160355599A1 (en) 2015-05-04 2016-12-08 Cytomx Therapeutics, Inc. Anti-cd71 antibodies, activatable anti-cd71 antibodies, and methods of use thereof
WO2017011618A1 (en) 2015-07-15 2017-01-19 The Curators Of The University Of Missouri Targeted nanoparticle conjugate and method for co-delivery of sirna and drug
US9644023B2 (en) 2013-12-09 2017-05-09 New York University Compositions and methods for phagocyte delivery of anti-staphylococcal agents
US20170174748A1 (en) 2014-03-20 2017-06-22 Bristol-Myers Squibb Company Serum albumin-binding fibronectin type iii domains
US20170348397A1 (en) 2016-06-03 2017-12-07 Janssen Biotech, Inc. Serum Albumin-Binding Fibronectin Type III Domains
US20190175651A1 (en) 2017-12-13 2019-06-13 Janssen Biotech, Inc. Immortalized car-t cells genetically modified to elminate t-cell receptor and beta 2-microglobulin expression
US20190184018A1 (en) 2007-12-04 2019-06-20 Alnylam Pharmaceuticals, Inc. Carbohydrate conjugates as delivery agents for oligonucleotides
US20190184028A1 (en) 2017-12-14 2019-06-20 Janssen Biotech, Inc. Targeting with firbronectin type iii like domain molecules
US20190256575A1 (en) 2014-04-08 2019-08-22 University Of Southern California Polypeptide compositions with type vii collagen fibronectin type iii-like repeats and treatment methods for wound closure and healing
US20190330361A1 (en) 2016-09-14 2019-10-31 Janssen Biotech, Inc. Chimeric Antigen Receptors Comprising BCMA-specific Fibronectin Type III Domains and Uses Thereof
US10597438B2 (en) * 2016-12-14 2020-03-24 Janssen Biotech, Inc. PD-L1 binding fibronectin type III domains
US10611823B2 (en) * 2016-12-14 2020-04-07 Hanssen Biotech, Inc CD137 binding fibronectin type III domains
US10626165B2 (en) 2016-12-14 2020-04-21 Janssen Biotech, Inc. CD8a-binding fibronectin type III domains

Patent Citations (195)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4281061A (en) 1979-07-27 1981-07-28 Syva Company Double antibody for enhanced sensitivity in immunoassay
US6018030A (en) 1986-11-04 2000-01-25 Protein Polymer Technologies, Inc. Peptides comprising repetitive units of amino acids and DNA sequences encoding the same
US6355776B1 (en) 1986-11-04 2002-03-12 Protein Polymer Technologies, Inc. Peptides comprising repetitive units of amino acids and DNA sequences encoding the same
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5643768A (en) 1989-10-05 1997-07-01 Optein, Inc. Cell-free synthesis and isolation of novel genes and polypeptides
US5658727A (en) 1991-04-10 1997-08-19 The Scripps Research Institute Heterodimeric receptor libraries using phagemids
US6172197B1 (en) 1991-07-10 2001-01-09 Medical Research Council Methods for producing members of specific binding pairs
US6582915B1 (en) 1991-12-02 2003-06-24 Medical Research Council Production of anti-self bodies from antibody segment repertories and displayed on phage
US6162903A (en) 1992-05-07 2000-12-19 Actinova Limited Immunoglobulin binding proteins derived from L protein and their uses
US5856456A (en) 1992-11-20 1999-01-05 Enzon, Inc. Linker for linked fusion polypeptides
US5643763A (en) 1994-11-04 1997-07-01 Genpharm International, Inc. Method for making recombinant yeast artificial chromosomes by minimizing diploid doubling during mating
WO1996038557A1 (en) 1995-06-02 1996-12-05 Genentech, Inc. Hepatocyte growth factor receptor antagonists and uses thereof
US5691157A (en) 1995-10-24 1997-11-25 The Research Foundation Of State University Of New York Method for detecting a mammal's prior exposure to radiation or radiomimetic chemicals
US5846456A (en) 1996-01-17 1998-12-08 National Science Council Method of making gradient index optical element
EP0985039A2 (en) 1997-06-12 2000-03-15 Research Corporation Technologies, Inc Artificial antibody polypeptides
US7119171B2 (en) 1997-06-12 2006-10-10 Research Corporation Technologies, Inc. Binding polypeptides
US7078490B2 (en) 1997-06-12 2006-07-18 Research Corporation Technologies, Inc. Artificial antibody polypeptides
US7153661B2 (en) 1997-06-12 2006-12-26 Research Corporation Technologies, Inc. Artificial antibody polypeptides
US6703199B1 (en) 1997-06-12 2004-03-09 Research Corporation Technologies, Inc. Artificial antibody polypeptides
US6673901B2 (en) 1997-06-12 2004-01-06 Research Corporation Technologies, Inc. Artificial antibody polypeptides
US6462189B1 (en) 1997-06-12 2002-10-08 Research Corporation Technologies Nucleic acids encoding artificial antibody polypeptides
US6521427B1 (en) 1997-09-16 2003-02-18 Egea Biosciences, Inc. Method for the complete chemical synthesis and assembly of genes and genomes
US6670127B2 (en) 1997-09-16 2003-12-30 Egea Biosciences, Inc. Method for assembly of a polynucleotide encoding a target polypeptide
US6846655B1 (en) 1998-06-29 2005-01-25 Phylos, Inc. Methods for generating highly diverse libraries
US7115396B2 (en) 1998-12-10 2006-10-03 Compound Therapeutics, Inc. Protein scaffolds for antibody mimics and other binding proteins
US20060270604A1 (en) 1998-12-10 2006-11-30 Compound Therapeutics, Inc. Pharmaceutical preparations of Fn3 polypeptides for human treatments
US20050255548A1 (en) 1998-12-10 2005-11-17 Phylos, Inc. Protein scaffolds for antibody mimics and other binding proteins
EP1137941A1 (en) 1998-12-10 2001-10-04 Phylos, Inc. Protein scaffolds for antibody mimics and other binding proteins
US20080015339A1 (en) 1998-12-10 2008-01-17 Dasa Lipovsek High affinity fibronectin derivatives
US20050038229A1 (en) 1998-12-10 2005-02-17 Dasa Lipovsek Protein scaffolds for antibody mimics and other binding proteins
US6818418B1 (en) 1998-12-10 2004-11-16 Compound Therapeutics, Inc. Protein scaffolds for antibody mimics and other binding proteins
US20060246059A1 (en) 1998-12-10 2006-11-02 Compound Therapeutics, Inc. Pharmaceutically acceptable FN3 polypeptides for human treatments
US6472147B1 (en) 1999-05-25 2002-10-29 The Scripps Research Institute Methods for display of heterodimeric proteins on filamentous phage using pVII and pIX, compositions, vectors and combinatorial libraries
EP1210428A1 (en) 1999-08-23 2002-06-05 Dana-Farber Cancer Institute, Inc. Pd-1, a receptor for b7-4, and uses therefor
WO2001014557A1 (en) 1999-08-23 2001-03-01 Dana-Farber Cancer Institute, Inc. Pd-1, a receptor for b7-4, and uses therefor
US8981063B2 (en) 1999-11-30 2015-03-17 Mayo Foundation For Medical Education And Research B7-H1 Antibodies
WO2001064942A1 (en) 2000-02-29 2001-09-07 Phylos, Inc. Protein scaffolds for antibody mimics and other binding proteins
EP1266025A1 (en) 2000-02-29 2002-12-18 Compound Therapeutics, Inc. Protein scaffolds for antibody mimics and other binding proteins
US20130226834A1 (en) 2000-04-27 2013-08-29 Networth Services, Inc. Systems and methods for determining the financial status of bonds
US7709214B2 (en) 2000-06-28 2010-05-04 Dana-Farber Cancer Institute, Inc. Methods for upregulating an immune response with agents that inhibit the intereaction between PD-L2 and PD-1
WO2002032925A2 (en) 2000-10-16 2002-04-25 Phylos, Inc. Protein scaffolds for antibody mimics and other binding proteins
US7794710B2 (en) 2001-04-20 2010-09-14 Mayo Foundation For Medical Education And Research Methods of enhancing T cell responsiveness
US20040259781A1 (en) 2001-08-09 2004-12-23 Ruth Chiquet-Ehrismann Anti-tumour agents and methods of identifying anti-tumour agents
US20040197332A1 (en) 2001-08-09 2004-10-07 Axel Ullrich Inhibitors of her3 activity
US6969108B2 (en) 2001-09-26 2005-11-29 Aisin Seiki Kabushiki Kaisha Vehicular door
US20050004029A1 (en) 2001-11-01 2005-01-06 Garcia Joe G.N. Methods and compositions for treating vascular leak using hepatocyte growth factor
WO2003104418A2 (en) 2002-06-06 2003-12-18 Research Corporation Technologies, Inc. Reconstituted polypeptides
US7842476B2 (en) 2002-09-06 2010-11-30 Isogenica Limited In vitro peptide expression library
WO2004029224A2 (en) 2002-09-30 2004-04-08 Compound Therapeutics, Inc. Methods of engineering spatially conserved motifs in polypeptides
WO2004058821A2 (en) 2002-12-27 2004-07-15 Domantis Limited Dual specific single domain antibodies specific for a ligand and for the receptor of the ligand
US20080241159A1 (en) 2003-04-16 2008-10-02 Genentech, Inc. Methods and compositions for selective modulation of vascularization
WO2005018534A2 (en) 2003-05-16 2005-03-03 Rosetta Inpharmatics, Llc Methods and compositions for rna interference
US7288638B2 (en) 2003-10-10 2007-10-30 Bristol-Myers Squibb Company Fully human antibodies against human 4-1BB
WO2005042708A2 (en) 2003-10-27 2005-05-12 Rosetta Inpharmatics Llc METHOD OF DESIGNING siRNAS FOR GENE SILENCING
US20070160533A1 (en) 2003-12-05 2007-07-12 Yan Chen Pharmacokinetic modulation and compositions for modified Fn3 polypeptides
US20070148126A1 (en) 2003-12-05 2007-06-28 Yan Chen Inhibitors of type 2 vascular endothelial growth factor receptors
US20080220049A1 (en) 2003-12-05 2008-09-11 Adnexus, A Bristol-Myers Squibb R&D Company Compositions and methods for intraocular delivery of fibronectin scaffold domain proteins
US20060040278A1 (en) 2004-01-27 2006-02-23 Cojocaru Gad S Novel nucleotide and amino acid sequences, and assays and methods of use thereof for diagnosis of ovarian cancer
US20050272083A1 (en) 2004-06-04 2005-12-08 Somasekar Seshagiri EGFR mutations
WO2007000671A2 (en) 2005-06-15 2007-01-04 Monoclonal Antibodies Therapeutics Anti-cd71 monoclonal antibodies and uses thereof for treating malignant tumour cells
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
US20070184476A1 (en) 2006-01-24 2007-08-09 Hui-Chu Hsieh Biomarkers for liver fibrotic injury
WO2007085815A2 (en) 2006-01-24 2007-08-02 Domantis Limited Ligands that bind il-4 and/or il-13
US20090311803A1 (en) 2006-03-31 2009-12-17 Way Jeffrey C Treatment Of Tumors Expressing Mutant EGF Receptors
US20150252097A1 (en) 2006-11-22 2015-09-10 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins for tyrosine kinases receptors, including igf-ir
WO2008079973A2 (en) 2006-12-21 2008-07-03 Centocor, Inc. Egfr binding peptides and uses thereof
US20120225870A1 (en) 2007-04-13 2012-09-06 Janne Pasi A Methods for treating cancer resistant to erbb therapeutics
WO2008127710A2 (en) 2007-04-13 2008-10-23 Dana Farber Cancer Institute Methods for treating cancer resistant to erbb therapeutics
US20110081345A1 (en) 2007-04-18 2011-04-07 Moore Margaret D Single chain fc, methods of making and methods of treatment
US20090042906A1 (en) 2007-04-26 2009-02-12 Massachusetts Institute Of Technology Methods for treating cancers associated with constitutive egfr signaling
WO2008156642A1 (en) 2007-06-15 2008-12-24 Vasgene Therapeutics, Inc. Non-immunoglobulin antigen binding scaffolds for inhibiting angiogenesis and tumor growth
US20110124527A1 (en) 2007-08-10 2011-05-26 Guido Cappuccilli Universal fibronectin type iii binding-domain libraries
WO2009023184A2 (en) 2007-08-10 2009-02-19 Protelix, Inc. Universal fibronectin type iii binding-domain libraries
US20090176654A1 (en) 2007-08-10 2009-07-09 Protelix, Inc. Universal fibronectin type III binding-domain libraries
WO2009058379A2 (en) 2007-10-31 2009-05-07 Medimmune, Llc Protein scaffolds
US20190184018A1 (en) 2007-12-04 2019-06-20 Alnylam Pharmaceuticals, Inc. Carbohydrate conjugates as delivery agents for oligonucleotides
US20110118144A1 (en) 2007-12-19 2011-05-19 Linus Hyun Engineered phage vectors for the design and the generation of a human non-antibody peptide or protein phage library via fusion to pix of m13 phage
WO2009086116A2 (en) 2007-12-19 2009-07-09 Centocor, Inc. Alternative scaffold protein fusions phage display via fusion to plx of m13 phage
WO2009085462A1 (en) 2007-12-19 2009-07-09 Centocor, Inc. Design and generation of human de novo pix phage display libraries via fusion to pix or pvii, vectors, antibodies and methods
JP2011507543A (en) 2007-12-27 2011-03-10 ノバルティス アーゲー Improved fibronectin-based binding molecules and their use
WO2009083804A2 (en) 2007-12-27 2009-07-09 Novartis Ag Improved fibronectin-based binding molecules and their use
WO2009102421A2 (en) 2008-02-14 2009-08-20 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins that bind egfr
JP2011517314A (en) 2008-02-14 2011-06-02 ブリストル−マイヤーズ スクイブ カンパニー Targeted therapeutics based on engineered proteins that bind to EGFR
US20110053842A1 (en) 2008-02-14 2011-03-03 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins that bind egfr
WO2009111691A2 (en) 2008-03-06 2009-09-11 Genentech, Inc. Combination therapy with c-met and egfr antagonists
WO2009126834A2 (en) 2008-04-11 2009-10-15 Galaxy Biotech, Llc Combination of hgf inhibitor and egf inhibitor to treat cancer
US20110038866A1 (en) 2008-05-02 2011-02-17 Novartis Ag Improved fibronectin-based binding molecules and uses thereof
CN102076713A (en) 2008-05-02 2011-05-25 诺瓦提斯公司 Improved fibronectin-based binding molecules and uses thereof
JP2011522517A (en) 2008-05-02 2011-08-04 ノバルティス アーゲー Improved fibronectin-based binding molecules and their use
WO2009133208A1 (en) 2008-05-02 2009-11-05 Novartis Ag Improved fibronectin-based binding molecules and uses thereof
US20090274693A1 (en) 2008-05-05 2009-11-05 Gilmer Tona M Method of Treating Cancer using a cMet and AXL Inhibitor and an ErbB Inhibitor
US20140349929A1 (en) 2008-05-22 2014-11-27 Bristol-Myers Squibb Company Multivalent fibronectin based scaffold domain proteins
US20090299040A1 (en) 2008-05-22 2009-12-03 Bristol-Myers Squibb Company Multivalent fibronectin based scaffold domain proteins
US20130012435A1 (en) 2008-05-22 2013-01-10 Bristol-Myers Squibb Company Multivalent fibronectin based scaffold domain proteins
JP2011520961A (en) 2008-05-22 2011-07-21 ブリストル−マイヤーズ スクイブ カンパニー Scaffold domain protein based on multivalent fibronectin
WO2009142773A2 (en) 2008-05-22 2009-11-26 Bristol-Myers Squibb Company Multivalent fibronectin based scaffold domain proteins
US8552154B2 (en) 2008-09-26 2013-10-08 Emory University Anti-PD-L1 antibodies and uses therefor
WO2010039248A1 (en) 2008-10-01 2010-04-08 Ludwig Institute For Cancer Research Methods for the treatment of cancer
US20100144601A1 (en) 2008-10-31 2010-06-10 Steven Jacobs Anti-TNF Alpha Fibronectin Type III Domain Based Scaffold Compositions, Methods and Uses
US20100216708A1 (en) 2008-10-31 2010-08-26 Steven Jacobs Fibronectin Type III Domain Based Scaffold Compositions, Methods and Uses
WO2010051274A2 (en) 2008-10-31 2010-05-06 Centocor Ortho Biotech Inc. Fibronectin type iii domain based scaffold compositions, methods and uses
WO2010051310A2 (en) 2008-10-31 2010-05-06 Centocor Ortho Biotech Inc. ANTI-TNFα FIBRONECTIN TYPE III DOMAIN BASED SCAFFOLD COMPOSITIONS, METHODS AND USES
US8278419B2 (en) 2008-10-31 2012-10-02 Centocor Ortho Biotech Inc. Fibronectin type III domain based scaffold compositions, methods and uses
US20120263723A1 (en) 2008-11-21 2012-10-18 Eli Lilly And Company c-Met Antibodies
WO2010060095A1 (en) 2008-11-24 2010-05-27 Bristol-Myers Squibb Company Bispecific egfr/igfir binding molecules
US20100179094A1 (en) 2008-11-24 2010-07-15 Bristol-Myers Squibb Company Bispecific egfr/igfir binding molecules
US8217149B2 (en) 2008-12-09 2012-07-10 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
US8741295B2 (en) 2009-02-09 2014-06-03 Universite De La Mediterranee PD-1 antibodies and PD-L1 antibodies and uses thereof
US8293482B2 (en) 2009-02-12 2012-10-23 Centocor Ortho Biotech Inc. Fibronectin type III domain based scaffold compositions, methods and uses
WO2010093627A2 (en) 2009-02-12 2010-08-19 Centocor Ortho Biotech Inc. Fibronectin type iii domain based scaffold compositions, methods and uses
US20100255056A1 (en) 2009-02-12 2010-10-07 Steven Jacobs Fibronectin Type III Domain Based Scaffold Compositions, Methods And Uses
WO2010115202A2 (en) 2009-04-03 2010-10-07 Dicerna Pharmaceuticals, Inc. Methods and compositions for the specific inhibition of kras by blunt ended double-stranded rna
US20100254989A1 (en) 2009-04-07 2010-10-07 Birgit Bossenmaier Bispecific Anti ErbB1 / Anti c Met Antibodies
WO2010115551A1 (en) 2009-04-07 2010-10-14 Roche Glycart Ag Bispecific anti-erbb-1/anti-c-met antibodies
WO2011005133A1 (en) 2009-07-07 2011-01-13 Siemens Aktiengesellschaft Apparatus and method for measuring multi-phase fluid flow
US20120270797A1 (en) * 2009-08-13 2012-10-25 Massachusetts Institute Of Technology Engineered proteins including mutant fibronectin domains
US8779108B2 (en) 2009-11-24 2014-07-15 Medimmune, Limited Targeted binding agents against B7-H1
US20130039927A1 (en) 2010-02-12 2013-02-14 University Of Rochester Antigenic mimics of discontinuous epitopes of pathogen recognized by broadly neutralizing antibodies
WO2011110642A2 (en) 2010-03-10 2011-09-15 Genmab A/S Monoclonal antibodies against c-met
WO2011130324A1 (en) 2010-04-13 2011-10-20 Medimmune, Llc Fibronectin type iii domain-based multimeric scaffolds
WO2011131746A2 (en) 2010-04-20 2011-10-27 Genmab A/S Heterodimeric antibody fc-containing proteins and methods for production thereof
US20110287009A1 (en) 2010-04-23 2011-11-24 Genentech, Inc. Production of Heteromultimeric Proteins
WO2011137319A2 (en) 2010-04-30 2011-11-03 Centocor Ortho Biotech Inc. Stabilized fibronectin domain compositions, methods and uses
US20110274623A1 (en) 2010-04-30 2011-11-10 Steven Jacobs Stabilized Fibronectin Domain Compositions, Methods and Uses
US8569227B2 (en) 2010-04-30 2013-10-29 Janssen Biotech, Inc. Stabilized fibronectin domain compositions, methods and uses
US20130184212A1 (en) 2010-05-26 2013-07-18 Bristol-Myers Squibb Company Fibronectin based scaffold proteins having improved stability
WO2011151412A1 (en) 2010-06-01 2011-12-08 Pierre Fabre Medicament Novel anti-cmet antibody
WO2012016245A2 (en) 2010-07-30 2012-02-02 Novartis Ag Fibronectin cradle molecules and libraries thereof
US20120244164A1 (en) 2010-09-30 2012-09-27 Ablynx N.V. Biological materials related to c-met
US20130273561A1 (en) 2010-10-29 2013-10-17 The Governing Council Of The University Of Toronto Lipid encapsulation of surface enhanced raman scattering (sers) nanoparticles
WO2012162418A1 (en) 2011-05-23 2012-11-29 The University Of North Carolina At Chapel Hill Methods and compositions for heterodimeric targeting ligands
US20120321666A1 (en) 2011-05-23 2012-12-20 Cooper Laurence J N T cell therapy for b cell lymphoma
US20120315639A1 (en) 2011-06-08 2012-12-13 Glenn Yaguang Deng Method and apparatus for single cell isolation and analysis
US20160041182A1 (en) 2011-09-27 2016-02-11 Janssen Biotech, Inc. Fibronectin Type III Repeat Based Protein Scaffolds with Alternative Binding Surfaces
US9897612B2 (en) 2011-09-27 2018-02-20 Janssen Biotech, Inc. Fibronectin type III repeat based protein scaffolds with alternative binding surfaces
WO2013049275A1 (en) 2011-09-27 2013-04-04 Janssen Biotech, Inc. Fibronectin type iii repeat based protein scaffolds with alternative binding surfaces
CN103827361A (en) 2011-09-27 2014-05-28 詹森生物科技公司 Fibronectin type iii repeat based protein scaffolds with alternative binding surfaces
US9200273B2 (en) 2011-09-27 2015-12-01 Janssen Biotech, Inc. Fibronectin type III repeat based protein scaffolds with alternative binding surfaces
US20140341917A1 (en) 2011-11-28 2014-11-20 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof
US9546368B2 (en) 2011-12-22 2017-01-17 Ionis Pharmaceuticals, Inc. Methods for modulating metastasis-associated-in-lung-adenocarcinoma-transcript-1 (MALAT-1) expression
US20140371296A1 (en) 2011-12-22 2014-12-18 Isis Pharmaceuticals, Inc. Methods for modulating metastasis-associated-in-lung-adenocarcinoma-transcript-1 (malat-1) expression
US20150005364A1 (en) 2012-01-05 2015-01-01 Bioneer Corporation High-efficiency nanoparticle-type double-helical oligo-rna structure and method for preparing same
US9326941B2 (en) 2012-01-05 2016-05-03 Bioneer Corporation High-efficiency nanoparticle-type double-helical oligo-RNA structure and method for preparing same
US9212224B2 (en) 2012-05-15 2015-12-15 Bristol-Myers Squibb Company Antibodies that bind PD-L1 and uses thereof
US20150118288A1 (en) 2012-05-23 2015-04-30 Ohio State Innovation Foundation Lipid Nanoparticle Compositions and Methods of Making and Methods of Using the Same
US9175082B2 (en) 2012-05-31 2015-11-03 Sorrento Therapeutics, Inc. Antigen binding proteins that bind PD-L1
US20150197571A1 (en) 2012-08-03 2015-07-16 Dana-Farber Cancer Institute, Inc. Single Agent Anti-PD-L1 and PD-L2 Dual Binding Antibodies and Methods of Use
US20150274835A1 (en) 2012-10-04 2015-10-01 Dana-Farber Cancer Institute, Inc. Human monoclonal anti-pd-l1 antibodies and methods of use
US20140155325A1 (en) 2012-11-21 2014-06-05 Janssen Biotech, Inc. Egfr and c-met fibronectin type iii domain binding molecules
US20140141000A1 (en) 2012-11-21 2014-05-22 Janssen Biotech, Inc. Bispecific EGFR/C-Met Antibodies
WO2014081944A2 (en) 2012-11-21 2014-05-30 Janssen Biotech, Inc. Egfr and c-met-fibronectin type iii domain binding molecules
WO2014081954A1 (en) 2012-11-21 2014-05-30 Janssen Biotech, Inc. BISPECIFIC EGFR/c-Met ANTIBODIES
US20140155326A1 (en) 2012-11-21 2014-06-05 Janssen Biotech, Inc. Egfr and c-met fibronectin type iii domain binding molecules
US20140255408A1 (en) 2012-11-21 2014-09-11 Janssen Biotech, Inc. Bispecific EGFR/C-Met Antibodies
WO2014100079A1 (en) 2012-12-21 2014-06-26 Merck Sharp & Dohme Corp. Antibodies that bind to human programmed death ligand 1 (pd-l1)
US20150355184A1 (en) 2012-12-21 2015-12-10 Robert H. Pierce Antibodies that bind to human programmed death ligand 1 (pd-l1)
EP2935329A1 (en) 2012-12-21 2015-10-28 Merck Sharp & Dohme Corp. Antibodies that bind to human programmed death ligand 1 (pd-l1)
US20140271467A1 (en) * 2013-03-12 2014-09-18 The Board Of Trustees Of The Leland Stanford Junior University Probes and methods of imaging non-hodgkins lymphoma
WO2014165082A2 (en) 2013-03-13 2014-10-09 Medimmune, Llc Antibodies and methods of detection
WO2014165093A2 (en) 2013-03-13 2014-10-09 Bristol-Myers Squibb Company Fibronectin based scaffold domains linked to serum albumin or a moiety binding thereto
WO2014189973A2 (en) 2013-05-20 2014-11-27 Genentech, Inc. Anti-transferrin receptor antibodies and methods of use
WO2014209804A1 (en) 2013-06-24 2014-12-31 Biomed Valley Discoveries, Inc. Bispecific antibodies
US20160303256A1 (en) 2013-09-30 2016-10-20 The University Of North Carolina At Chapel Hill Methods and compositions for self-assembly system of nanoparticles and microparticles for multi-targeting specificity
US20190263915A1 (en) 2013-10-14 2019-08-29 Janssen Biotech, Inc. Cysteine engineered fibronectin type iii domain binding molecules
US20150104808A1 (en) 2013-10-14 2015-04-16 Janssen Biotech, Inc. Cysteine Engineered Fibronectin Type III Domain Binding Molecules
US10196446B2 (en) 2013-10-14 2019-02-05 Janssen Biotech, Inc. Cysteine engineered fibronectin type III domain binding molecules
WO2015061668A1 (en) 2013-10-25 2015-04-30 Dana-Farber Cancer Institute, Inc. Anti-pd-l1 monoclonal antibodies and fragments thereof
US9644023B2 (en) 2013-12-09 2017-05-09 New York University Compositions and methods for phagocyte delivery of anti-staphylococcal agents
WO2015092393A2 (en) 2013-12-17 2015-06-25 Kymab Limited Human targets
WO2015109124A2 (en) 2014-01-15 2015-07-23 Kadmon Corporation, Llc Immunomodulatory agents
US20150203580A1 (en) 2014-01-23 2015-07-23 Regeneron Pharmaceuticals, Inc. Human Antibodies to PD-L1
US20170174748A1 (en) 2014-03-20 2017-06-22 Bristol-Myers Squibb Company Serum albumin-binding fibronectin type iii domains
US20190256575A1 (en) 2014-04-08 2019-08-22 University Of Southern California Polypeptide compositions with type vii collagen fibronectin type iii-like repeats and treatment methods for wound closure and healing
US20150346208A1 (en) 2014-05-29 2015-12-03 Spring Bioscience Corporation PD-L1 Antibodies and Uses Thereof
WO2015195163A1 (en) 2014-06-20 2015-12-23 R-Pharm Overseas, Inc. Pd-l1 antagonist fully human antibody
WO2016004043A1 (en) 2014-06-30 2016-01-07 Blend Therapeutics, Inc. Targeted conjugates and particles and formulations thereof
WO2016000619A1 (en) 2014-07-03 2016-01-07 Beigene, Ltd. Anti-pd-l1 antibodies and their use as therapeutics and diagnostics
WO2016086021A1 (en) 2014-11-25 2016-06-02 Bristol-Myers Squibb Company Novel pd-l1 binding polypeptides for imaging
US20170258948A1 (en) 2014-11-25 2017-09-14 Bristol-Myers Squibb Company Novel pd-l1 binding polypeptides for imaging
WO2016086036A2 (en) 2014-11-25 2016-06-02 Bristol-Myers Squibb Company Methods and compositions for 18f-radiolabeling of biologics
US20160355599A1 (en) 2015-05-04 2016-12-08 Cytomx Therapeutics, Inc. Anti-cd71 antibodies, activatable anti-cd71 antibodies, and methods of use thereof
US20190202927A1 (en) 2015-05-04 2019-07-04 Cytomx Therapeutics, Inc. Anti-cd71 antibodies, activatable anti-cd71 antibodies, and methods of use thereof
US20160326232A1 (en) 2015-05-06 2016-11-10 Janssen Biotech, Inc. Prostate Specific Membrane Antigen Binding Fibronectin Type III Domains
WO2016179534A2 (en) 2015-05-06 2016-11-10 Janssen Biotech, Inc. Prostate specific membrane antigen binding fibronectin type iii domains
WO2016197071A1 (en) 2015-06-05 2016-12-08 New York University Compositions and methods for anti-staphylococcal biologic agents
US20190127444A1 (en) 2015-06-05 2019-05-02 New York University Compositions and methods for anti-staphylococcal biologic agents
WO2017011618A1 (en) 2015-07-15 2017-01-19 The Curators Of The University Of Missouri Targeted nanoparticle conjugate and method for co-delivery of sirna and drug
CN105907719A (en) 2016-04-18 2016-08-31 李华顺 Anti BOBO1 CAR-T cell and preparation and application thereof
US20170348397A1 (en) 2016-06-03 2017-12-07 Janssen Biotech, Inc. Serum Albumin-Binding Fibronectin Type III Domains
US20190330361A1 (en) 2016-09-14 2019-10-31 Janssen Biotech, Inc. Chimeric Antigen Receptors Comprising BCMA-specific Fibronectin Type III Domains and Uses Thereof
US10597438B2 (en) * 2016-12-14 2020-03-24 Janssen Biotech, Inc. PD-L1 binding fibronectin type III domains
US10611823B2 (en) * 2016-12-14 2020-04-07 Hanssen Biotech, Inc CD137 binding fibronectin type III domains
US10626165B2 (en) 2016-12-14 2020-04-21 Janssen Biotech, Inc. CD8a-binding fibronectin type III domains
US20190175651A1 (en) 2017-12-13 2019-06-13 Janssen Biotech, Inc. Immortalized car-t cells genetically modified to elminate t-cell receptor and beta 2-microglobulin expression
US20190184028A1 (en) 2017-12-14 2019-06-20 Janssen Biotech, Inc. Targeting with firbronectin type iii like domain molecules

Non-Patent Citations (161)

* Cited by examiner, † Cited by third party
Title
Adjei et al., "Early Clinical Development of ARQ197, a Selective, Non-ADP-Competitive Inhibitor Targeting MET Tyrosine Kinase for the Treatment of Advanced Cancers," The Oncologist, vol. 16, pp. 788-799 (2011).
Alderson et al., "Molecular and Biological Characterization of Human 4-1 BB and its Ligand", Eur. J_ Immunol., vol. N, pp. 2219-2227, 1994.
Alfthan et al., "Properties of a single-chain antibody containing different linker peptides," Protein Engineering, vol. B, No. 7, pp. 725-731 (1995).
Attwood TK. Genomics. The Babel of bioinformatics. Science. 290(5491 ):471-473, 2000.
Basel Ga et al., "Critical Update and Emerging Trends in Epidermal Growth Factor Receptor Targeting in Cancer," Journal of Clinical Oncology, vol. 23, No. 11, pp. 2445-2459 (2005).
Bass, et al., "Hormone Phage: An Enrichment Method for Variant Proteins with Altered Binding Properties," PROTEINS: Structure, Function, and Genetics, 8: 309-314 (1990).
Batley et al., "Inhibition of FGF-1 Receptor Tyrosine Kinase Activity By PD 161570, a New Protein-Tyrosine Kinase nhibitor," Life Sciences, vol. 62, No. 20, pp. 143-150 (1998).
Bean et al., "MET amplification occurs with or without T790M mutations in EGFR mutant lung tumors with acquired esistance to gefilinib or erlotinib," Proceedings of the National Academy of Science, vol. 104, No. 52, pp. ) 0932-20937 (2007).
Binz et al., "High-affinity binders selected from designed ankyrin repeat protein libraries," Nature Biotechnology, vol. e2, No. 5, pp. 575-582 (May 2004).
Binz, et al., "Engineered proteins as specific binding reagents," Current Opinion in Biotechnology, 16:459-469 (2005).
Birtalan et al., "The Intrinsic Contributions of Tyrosine, Serine, Glycine and Arginine to the Affinity and Specificity of Antibodies," Journal of Molecular Biology, vol. 377, pp. 1518-1528 (2008).
Bork et al., "Proposed acquisition of an animal protein domain by bacteria," Proceedings of the National Academy of Science, USA, vol. 89, pp. 8990-8994 (1992).
Brown et al., "Tolerance of single, but not multiple, amino acid replacements in antibody VH CDR 2: a means of minimizing B cell wastage from somatic hypermutation", J. Immuno. (1996) pp. 3285-3291.
Burgess et al., "Possible dissociation of the heparin-binding and mitogenic activities of heparin-binding (acidic fibroblast) growth factor-1 from its receptor-binding activities by site-directed mutagenesis of a single lysine residue" J Cell Biol (1990) 111:pp. 2129-2138.
Burton Earle Barnett et al: "Disclosures", Blood, vol. 128, No. 22, Dec. 2, 2016 (Dec. 2, 2016), pp. 4557-4557, XP055711182, US ISSN: 0006-4971, doi: 10.1182/blood.V128.22.4557.4557 *abstract*.
BURTON EARLE BARNETT, XINXIN WANG, DAVID L. HERMANSON, YENING TAN, ERIC M. OSERTAG, DEVON J. SHEDLOCK: "Disclosures", BLOOD, AMERICAN SOCIETY OF HEMATOLOGY, US, vol. 128, no. 22, 2 December 2016 (2016-12-02), US , pages 4557 - 4557, XP055711182, ISSN: 0006-4971, DOI: 10.1182/blood.V128.22.4557.4557
C.N. Pace, "Determination and Analysis of Urea and Guanidine Hydrochloride Denaturation Curves," Methods in Enzymology, 131: 266-280 (1986).
Capellas, "Enzymatic Condensation of Cholecystokinin CCK-8 (4-6) and CCK-8 (7-8) Peptide Fragments in Organic Media", Biotechnology and Bioengineering (1997) vol. 56, No. 4, pp. 456-463.
Cappuzzo et al., "Epidermal Growth Factor Receptor Gene and Protein and Gefilinib Sensitivity in Non-small-Cell ung Cancer," Journal of the National Cancer Institute, vol. 97, pp. 643-655 (2005).
Chen et al., "Cell-Surface Display of Heterologous Proteins: From High-Throughput Screening to Environmental Applications", Biotechnology and Bioengineering, (2002) vol. 79, No. 5, pp. 496-503.
Chiba et al., Amyloid Fibril Formation in the Context of Full-length Protein Effects of Praline mutations on the Amyloid fibril formation of b2-Microglobulin, Journal of Biological Chemistry, vol. 278, No. 47, pp. 47016-47024, Nov. 2003.
Chimu Ra et al., "Expression of c-mel/HGF Receptor in Human Non-small Cell Lung Carcinomas in vitro and in vivo and Its Prognostic Significance," Japan Journal of Cancer Research, vol. 87. pp. 1063-1069 (1996).
Christensen et al., "c-Met as a target for human cancer and characterization of inhibitors for therapeutic ntervention," Cancer Letters, vol. 225, pp. 1-26 (2005).
Clarke, et al., "Folding and Stability of a Fibronectin Type III Domain of Human Tenascin," Journal of Molecular Biology, 270: 771-778 (1997).
Cooper et al., "4-1 BB (CD 137) controls the clonal expansion and survival of COB T cells in vivo but does not t: ontribute the development of cytotoxicity", Eur. J_ Immunol., vol. 32, pp. 521-529, 2002.
Cooper et al., "Molecular cloning of a new transforming gene from a chemically transformed human cell line," Nature, vol. 311, pp. 29-33 (1984).
Cota, et al., "Two Proteins with the Same Structure Respond very Differently to Mutation: The Role of Plasticity in Protein Stability", Journal of Molecular Biology, 302, 713-725 (2000).
DeBenedette et al., "Role of 4-1BB Ligand in Costimulation of T Lymphocyte Growth and its Upregulation on M12 B rmphomas by cAMP," J_ Exp_ Med., vol. 181, pp. 985-992 (1995).
Dehouck, et al., "Fast and accurate predictions of protein stability changes upon mutations using statistical potentials and neural networks: PoPMuSiC-2.0," Bioinformatics, 25(19): 2537-2543 (2009).
DeRoock et al., "Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis," Lancet Oncology, vol. 11, pp. 753-762 (2010).
Diem et al., "Selection of high-affinity Centyrin FN3 domains from a simply library diversified at a combination of strand and loop positions." Protein Engin Design (2014) Selection 27(10): 419-429.
Dineen, et al., "The Adnectin CT-322 is a novel VEGF receptor 2 inhibitor that decreases tumor burden in an orthotopic mouse model of pancreatic cancer," BMC Cancer, 8: 352-361 (2008).
Downward et al., "Autophosphorylation sites on the epidermal growth factor receptor," Nature, vol. 311, pp. 183-485 ( 1984).
Dutta, et al., "High-affinity fragment complementation of a fibronectin type III domain and its application to stability enhancement," Protein Science, 14: 2838-2848 (2005).
Engelman et al., "MET Amplification Leads to Gefitinib Resistance in Lung Cancer by Activating ERBB3 Signaling," Science, vol. 316, pp. 1039-1043 (2007).
Ferguson, Kathryn M., "Structure-Based View of Epidermal Growth Factor Receptor Regulation," Annual Review of Biophysics, vol. 37, pp. 535-373 (2008).
Final Office Action dated Jul. 10, 2020 in U.S. Appl. No. 15/637,276.
Final Office Action dated Jul. 21, 2020 in U.S. Appl. No. 16/218,990.
Garcia-Ibilcieta, et al., "Simple method for production of randomized human tenth fibronectin domain III libraries for use in combinatorial screening procedures," Bio Technologies, 44: 559-562 (2008).
Garon et al., "Pembrolizumab for the Treatment of Non-Small-Cell Lung Cancer," The New England Journal of Medicine, vol. 372, No. 21, pp. 2018-2028 (May 21, 2015).
Garrard, et al., "Selection of an anti-IGF-1 Fab from a Fab phage library created by mutagenesis of multiple CDR loops," Gene, 128: 103-109 (1993).
GenBank Accession No. NP 001120972, Apr. 20, 2020.
GenBank Accession No. NP_002151, May 9, 2020.
Getmanova, et al., "Antagonists to Human and Mouse Vascular Endothelial Growth Factor Receptor 2 Generated by Directed Protein Evolution In Vitro," Chemistry & Biology, 13: 549-556 (2006).
Gill et al., "Monoclonal Anti-epidermal Growth Factor Receptor Antibodies Which Are Inhibitors of Epidermal Growth racier Binding and Antagonists of Epidermal Growth Factor-stimulated tyrosine Protein Kinase Activity," The Journal Jf Biological Chemistry, vol. 259, No. 12, pp. 7755-7760 (1984).
Goldberg et al., "Engineering a Targeted Delivery Platform using Centyrins" Protein Engineering, Design & selection, vol. 29, No. 12, pp. 563-572, 2016.
Goldstein et al., "Biological efficacy of a chimeric antibody to the epidermal growth factor receptor in a human umor xenografl model," Clinical Cancer Research, vol. 1, pp. 1311-1318 (1995).
Gramaglia et al., "Co-stimulation of antigen-specific CD4T cells by 4-1BB ligand," Eur. J. Immunol., vol. 30, pp. ô €?″92-402 (2000).
Grünwald et al., "Developing Inhibitors of the Epidermal Growth Factor Receptor for Cancer Treatment," Journal of he National Cancer Institute, vol. 95, No. 12, pp. 851-867 (2003).
Hackel et al. Use of 64Cu-labeled fibronectin domain with EGFR-overexpressing tumor xenograft: molecular imaging. Radiol 263(1) : 179-188, 2012. *
Hackel, et al., "Picomolar Affinity Fibronectin Domains Engineered Utilizing Loop Length Diversity, Recursive Mutagenesis, and Loop Shuffling," Journal of Molecular Biology, 381: 1238-1252 (2008).
Hackel, et al., "Stability and CDR Composition Biases Enrich Binder Functionality Landscapes," Journal of Molecular Biology, 401: 84-96 (2010).
Hallewell et al., "Genetically Engineered Polymers of Human CuZN Superoxide Dismutase," The Journal of Biological Chemistry, vol. 264, No. 9, pp. 5260-5268 (1989).
Hamill et al., "The Effect of Boundary Selection on the Stability and Folding of the Third Fibronectin Type III Domain from Human Tenascin", Biochemistry, 37: 8071-8079 (1998).
Hanes et al, "In vitro selection and evolution of functional proteins by using ribosome display," Proceedings of the National Academy of Sciences USA, vol. 94, pp. 4937-4942 (1997).
Helms et al. Destabilizing loop swaps in the CDRs of an immunoglobulin VL domain. Protein Science 4:2073-2081, 1995.
Hirsch et al, "Combination of EGFR gene copy number and protein expression predicts outcome for advanced non-, mall-cell lung cancer patients treated with gefitnib," Annals of Oncology, vol. 18, pp. 752-760 (2007).
Hoogenboom et al., "Natural and designer binding sites made by phage display technology" Immunology Today (2000) vol. 21, No. 8, pp. 371-378.
Hurtado et al., "Potential role of 4-1 BB in T cell Activation Comparison with the Costimulatory Molecule CD28", Journal of Immunology, vol. 155, pp. 3360-3367, 1995.
Hurtado et al., "Signals through 4-1BB are Costimulatory to previously activated splenic T cells and inhibit activation-induced cell death", Journal of Immunology, vol. 158, pp. 2600-2609, 1997.
Hylarides et al., "Preparation and in Vivo Evaluation of an N-9p-[1251]1odophenethyl) maleimide—Antibody Conjugate" Bioconjugate Chem., vol. 2, pp. 435-440, 1991.
Hynes et al., "ERBB Receptors and Cancer: the Complexity of Targeted Inhibitors," Nature Reviews, vol. 5, pp. 341-356 (2005).
Itoh, et al., "Application of Inverse Substrates to Trypsin-Catalyzed Peptid Synthesis", Bioorganic Chemistry (1996) 24, 0007, pp. 59-68.
Jacobs et al., "Design of novel FN3 domains with high stability by a consensus sequence approach," Protein Engineering, Design & Selection, vol. 25, No. 3, pp. 107-117 (2012).
Jacobs et al., "FN3 Domain Engineering", Protein Engineering, pp. 145-162, 2012.
Jacobs et al., "Fusion to a highly stable consensus albumin binding domain allows for tunable pharmacokinetics", Protein Engineering, Design & Selection, vol. 28, No. 10, pp. 385-393, 2015.
Jänne et al., "Effect of Epidermal Growth Factor Receptor Tyrosine Kinase Domain Mutations on the Outcome of Patients with Non-small Cell Lung Cancer Treated with Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors," Clinical Cancer Research, vol. 12, No. 14 Suppl, pp. 4416s-4420s (2006).
Karatan, et al., "Molecular Recognition Properties of FN3 Mono bodies that Bind the Src SH3 Domain," Chemistry & Biology, 11: 835-844(2004).
Klein et al. "Abstract LB-312: Bispecific Centyrin Simultaneously targeting EGFR and c—Met demonstrates improved ô €?'ctivity compared to the mixture of single agents", Cancer Research, 73 (8 Supplement), Abstract LB-312, Apr. 2013.
Knappik, et al., "Fully Synthetic Human Combinatorial Antibody Libraries (HuCAL) Based on Modular Consensus Frameworks and CDRs Randomized with Trinucleotides," Journal of Molecular Biology, 296: 57-86 (2002).
Koide et al., "High-affinity single-domain binding proteins with a binary-code interface," PNAS, vol. 104, No. 16, pp. 6632-6637(Apr. 17, 2017).
Koide, et al., "The Fibronectin Type III Domain as a Scaffold for Novel Binding Proteins," Journal of Molecular Biology, 284: 1141-1151 (1998).
Koide, et al., Teaching an Old Scaffold New Tricks: Monobodies Constructed Using Alternative Surfaces of the FN3 Scaffold, Journal of Molecular Biology, 415: 393-405 (2012).
Koivunen et al. Identification of Receptor Ligands with Phage Display Peptide Libraries J Nucl Med; 40:883-888, 1999.
Kumaran et al., "Confrmationally driven protease-catalyzed splicing of peptide segments: V8 protease-mediated syntheses of fragments derived from thermolysin and ribonuclease A", Protein Science, (1997) 6: pp. 2233-2241.
Kunkel et al., "Rapid and Efficient Site-Specific Mutagenesis without Phenotypic Selection", Methods In Enzymology, (1987) vol. 154 pp. 367-375.
Kuntz. Structure-based strategies for drug design and discovery. Science. 1992 257(5073):1078-1082.
Kwon et al., cDNA sequences of two inducible T-cell genes, Proc. Natl. Acad. Sci., vol. 86, pp. 1963-1967, Mar. 1989.
Langstein et al., "CD137 (ILA/4-1 BB), a Member of the TNF Receptor Family, Induces Monocyte Activation via Bidirectional Signaling," The Journal of Immunology, vol. 160, pp. 2488-2494 (1998).
Langstein et al., "CD137 Induces Proliferation and Endomitosis in Monocytes," Blood, vol. 94, No. 9, pp. 3161-3168 1999).
Langstein et al., Identification of CD137 as a potent monocyte survival factor, Journal of Leukocyte Biology, vol. 65, pp. 829-833, Jun. 1999.
Lazar et al., "Transforming growth factor alpha: mutation of aspartic acid 47 and leucie 48 results in different biological activities", Mol Cell Biol. (1988) 8: pp. 1247-1252.
Lee et al., "4-1BB Promotes the Survival of COB+ T Lymphocytes by Increasing Expression of Bcl-xL and Bfl-11," The Journal of Immunol., vol. 169, pp. 4882-4888 (2002).
Lee et al., "A Glu-ruea-Lys Ligand-conjugated Lipid nanoparticle/siRNA System Inhibits Androgen Receptor Expression In Vivo", Molecular Therapy-Nucleic Acids (2016) 5, e348: pp. 1-11.
Lehmann et al., Engineering proteins for thermostability the use of sequence alignments versus rational design and directed evolution, Current Opinion in Biotechnology, vol. 12, pp. 371-375 (2001).
Lejon et al., "Structural basis for the binding of naproxen to human serum albumin in the presence of fatty acids and the GA module", Acta Cryst. (2008) F pp. 64-69.
Lepenies et al., "The Role of Negative Costimulators Dunng Parasitic Infections," Endocrine, Metabolic & Immune Disorders—Drug Targets, vol. 8, pp. 279-288 (2008).
Li et al., "Skin toxicities associated with epidermal growth factor receptor inhibitors," Target Oncology, vol. 4, pp. 107-119 (2009).
Linardou et al., "Somatic EGFR mutations and efficacy of tyrosine kinase inhibitors in NSCLC," National Review of :; linical Oncology, vol. 6, pp. 352-366 (2009).
Lipovsek, et al., "Evolution of an Interloop Disulfide Bond in High-Affinity Antibody Mimics Based on Fibronectin Type III Domain and Selected by Yeast Surface Display: Molecular Convergence with Single-Domain Camelid and Shark Antibodies," Journal of Molecular Biology, 368: 1024-1041 (2007).
Lohse et al., Fluorescein-Conjugated Lysine monomers for Solid Phase Synthesis of Fluorescent Peptides and PNA Pligomers Bioconjugate Chem, vol. 8, pp. 503-509, 1997 .pdf.
Ma et al., "c-Met: Structure, functions and potential for therapeutic inhibition," Cancer and Metastasis Reviews, vol. 22 pp. 309-325 (2003).
Määttä et al., "Proteolytic Cleavage and Phosphorylation of a Tumor-associated ErbB4 Isoform Promote Ligand-ndependent Survival and Cancer Cell Growth," Molecular Biology, vol. 17, pp. 67-79 (2006).
Makkouk Amani et al: "Rationale for anti-CD137 cancer immunotherapy", European Journal of Cancer, Elsevier, Amsterdam, NL, vol. 54, Jan. 2, 2016 (Jan. 2, 2016), pp. 112-119, XP029401784, ISSN: 0959-8049, DOI: 10.1016/j.ejca.2015.09.026 *abstract**p. 114, right-hand column, paragraph 4—p. 116, right-hand column, paragraph 1**table 1*.
MAKKOUK AMANI; CHESTER CARIAD; KOHRT HOLBROOK E.: "Rationale for anti-CD137 cancer immunotherapy", EUROPEAN JOURNAL OF CANCER, ELSEVIER, AMSTERDAM NL, vol. 54, 2 January 2016 (2016-01-02), Amsterdam NL , pages 112 - 119, XP029401784, ISSN: 0959-8049, DOI: 10.1016/j.ejca.2015.09.026
Mamluk et al., "Anti-tumor effect of CT-322 as an Adnectin inhibitor of vascular endothelial growth factor receptor-2", mAbs, 2(2), pp. 199-208, 2010.
Mattheakis et al., "An in vitro polysome display system for identifying ligands from very large peptide libraries", Proc. Natl. Acad. Sci. (1994) Vo.. 91, pp. 9022-9026.
Maus et al., Ex vivo expansion of polyclonal and antigen-specific cytotoxic T lymphocytes by artificial APCs Expressing ligands for the T-cell receptor, CD28 and 4-1BB Nature Biotechnology, vol. 20, pp. 143-148, Feb. 2002.
McCracken, M.N. (2014) Non-invasive monitoring of hematopoietic reconstitution and immune cell function through Positron Emission Tomography. (Doctoral dissertation, University of California, Los Angeles). Retrieved from Proquest Dissertations and Theses. (Thesis No. 3633653; ProQuest Document ID 1611185390). *
McLaughlin et al., "Quantitative Assessmenet of the Heterogeneity of PD-L 1 Expression in Non-small Cell Lung Dancer (NSCLC)," JAMA Oncol., vol. 2, No. 1, pp. 46-54, (Jan. 2016).
Meinke et al., "Cellulose-Binding Polypeptides from Cellulomonas fimi: Endoglucanase D (CenD), a Family A b-1,4-Glucanase," Journal of Bactenology, vol. 175, No. 7, pp. 1910-1918 (1993).
Mendelsohn et al., "Epidermal Growth Factor Receptor Targeting in Cancer," Seminars in Oncology, vol. 33, pp. 369-385 (2006).
Mendelsohn et al., "The EGF receptor family as targets for cancer therapy," Oncogene, vol. 19, pp. 6550-6565 2000).
Michel et al., "A soluble form of CD137 (ILA/4-1BB), a member of the TNF receptor family, is released by activated ymphocytes and is detectable in sera of patients with rheumatoid arthritis," Eur. J_ Immunol., vol. 28, pp. 290-295 1998).
Michel et al., "CD137-induced apoptosis is independent of CD95," Immunology, vol. 98, pp. 42-46 (1999).
Michel et al., "Expression of soluble CD137 correlates with activation-induced cell death of lymphocytes", Cytokine, vol. 12, No. 6, pp. 742-746, 2000.
Miller et al Ligand binding to proteins: the binding landscape model. Protein Sci. Oct. 1997;6(10):2166-79.
Natarajan et al. A novel engineered anti-CD20 tracer enables early time PET imaging in a humanized transgenic mouse model of B-cell Non-Hodgkins lymphoma. Clin Cancer Res 19(24): 6820-6829, 2013. *
NCBI Reference Sequence NP _005219.2, "Epidermal Growth Factor Receptor Isoform a Precursor [Homo sapiens]," pp. 1-14 (May 18, 2014).
Non-Final Office Action dated Feb. 3, 2021 in U.S. Appl. No. 16/218,990.
Non-Final Office Action dated Jul. 9, 2021 in U.S. Appl. No. 16/821,064.
Notice of Allowance dated Mar. 3, 2020 in U.S. Appl. No. 15/840,303.
Odegrip et al., "CIS display: In vitro selection of peptides from libraries of protein-DNA complexes," Proceedings of he National Academy of Science USA, vol. 101, No. 9, pp. 2806-2810 (2004).
Olson et al., "Design, expression, and stability of a diverse protein library based on the human fibronectin type III ô €,?omain," Protein Science, vol. 16, pp. 476-484 (2007).
Pace, "Determination and Analysis of Urea and Guanidine Hydrochloride Denaturation Curves", Methods in Enzymology (1986) vol. 131, pp. 266-280.
Panek et al.,"In Vitro Pharmacological Characterization of PD 166285, a New Nanomolar Potent and Broadly Active Protein Tyrosine Kinase Inhibitor," The Journal of Pharmacology and Experimental Therapeutics, vol. 283, No. 3, pp. 1433-1444 (1997).
Parker, et al., "Antibody mimics based on human fibronectin type three domain engineered for thermostability and high-affinity binding to vascular endothelial growth factor receptor two," Protein Engineering, Design & Selection, 18(9):435-444 (2005).
Pauly et al., CD137 is expressed by follicular dendritic cells and costimulates B lymphocyte activation in germinal t; enters. Journal of Leukocyte Biology, vol. 72, pp. 35-42, Jul. 2002.
Peters et al., "MET: a promising anticancer therapeutic target," Nature Reviews Clinical Oncology, vol. 9, pp. 314-326 (2012).
Prewett et al., "Mouse-Human chimeric Anti-Epidermal Growth Factor Receptor Antibody C225 Inhibits the Growth Jf Human Renal Cell Carcinoma Xenografts in Nude Mice," Clinical Cancer Research, vol. 4, pp. 2957-2966 (1998).
Reiss et al. Inhibition of platelet aggregation by grafting RGD and KGD sequences on the structural scaffold of small disulfide-rich proteins. Platelets 17(3):153-157, 2006.
Riel Yet al., "Clinical Course of Patients with Non-Small Cell Lung Cancer and Epidermal Growth Factor Receptor Exon 19 and Exon 21 Mutations Treated with Gefitinib or Erlotinib," Clinical Cancer Research, vol. 12, No. 3, pp. g39-844 (2006).
Roberts et al., "RNA-peptide fusions for the in vitro selection of peptides and proteins," Proceedings of the National Academy of Science USA, vol. 94, pp. 12297-12302 (1997).
Robinson et al., "Covalent Attachment of Arc Repressor Subunits by a Peptide Linker Enhances Affinity for Operator DNA," Biochemistry, vol. 35, pp. 109-116 (1996).
Rudikoff el al., "Single amino acid substitution altering antigen-binding specificity", Proc Natl Acad Sci (1982) 79(6): pp. 1979-1983.
Rybalov et al., "PSMA, EpCAM, VEGF and GRPR as Imaging Targets in Locally Recurrent Prostate Cancer after Radiotherapy", Int. J. Mol. Sci. (2014) 15, pp. 6046-6061.
Sakakura et al., "Gains, Losses, and Amplifications of Genomic Materials in Primary Gastric Cancers Analyzed by :; omparative Genomic Hybridization," Genes, Chromosomes & Cancer, vol. 24, pp. 299-305 (1999).
Schmidt et al., "Novel mutations of the MET proto-0ncogene in papillary rental carcinomas," Oncogene, vol. 18, pp. ]343-2350 (1999).
Schwarz et al., "ILA, a Member of the Human Nerve Growth FactorfTumor Necrosis Factor Receptor Family, Regulates T-Lymphocyte Proliferation and Survival," Blood, vol. 87, No. 7, pp. 2839-2845 (Apr. 1, 1996).
Shalom D. Goldberg et al: "Engineering a targeted delivery platform using Centyrins", Protein Engineering, Design and Selection, Oct. 13, 2016 (Oct. 13. 2016), XP055384705, GB ISSN: 1741-0126, DOI: 10.1093/protein/gzw054 *abstract**p. 564, left-hand column, paragraph 2—right-hand column, line 3** p. 567, right-hand column, paragraph 2**p. 568, right-hand column, paragraph 2—p. 569, left-hand column, paragraph 2**table l**figure 1a*.
SHALOM D. GOLDBERG, ROSA M.F. CARDOSO, TRICIA LIN, TRACY SPINKA-DOMS, DONNA KLEIN, STEVEN A. JACOBS, VADIM DUDKIN, GARY GILLILAND,: "Engineering a targeted delivery platform using Centyrins", PROTEIN ENGINEERING, DESIGN AND SELECTION, OXFORD JOURNAL, LONDON, GB, GB , XP055384705, ISSN: 1741-0126, DOI: 10.1093/protein/gzw054
Shuford et al., "4-18B Costimulatory Signals Preferentially Induce COB+ T Cell Proliferation and Lead to the amplification In Vivo of Cytotoxic T Cell Responses," J_ Exp_ Med., vol. 186, No. 1, pp. 47-55 (Jul. 7, 1997).
Siegfried et al.,"The Clinical Significance of Hepatocyte Growth Factor for Non-Small Cell Lung Cancer," Annals of Thoracic Surgery, vol. 66, pp. 1915-1918 (1998).
Sierra et al., "c-MET as a potential therapeutic target and biomarker in cancer," Therapeutic Advances in Medical :: >ncology, vol. 3, No. 51, pp. 521-535 (2011).
Siggers et al. Conformational dynamics in loop swap mutants of homologous fibronectin type III domains. Biophys J. Oct. 1, 2007 ;93(7):2447-56.
Skerra, et al., "Engineered protein scaffolds for molecular recognition," Journal of Molecular Recognition, 13: 167-187 (2000).
Skolnick et al. From genes to protein structure and function: novel applications of computational approaches in the genomic era. Trends Biotechnol. 18(1 ):34-9, 2000.
Slonomics® Technology Website "https://www.morphosys.com/science/drug-development-capabilities/slonomics", May 12, 2020.
Smith, "Filamentous fusion phage: novel expression vectors that display cloned antigens on the virion surface. ", Association of Science (1985) vol. 228, pp. 1315(3).
Song et al. Cancer stem cells—an old idea that's new again: implications for the diagnosis and treatment of breast cancer. Expert Opin Biol Ther 7:4):431-438, 2007.
Stamos et al., "Crystal structure of the HGF b-chain in complex with the Serna domain of the Met receptor," The EMBO Journal, vol. 23, pp. 2325-2335 (2004).
Steiner, et al., "Efficient Selection of DARPins with Sub-nonomolar Affinities using SRP Phage Display," Journal of Molecular Biology, 382: 1211-1227 (2008).
Strand et al., "Site-Specific Radioiodination of HER2-Targeting Affibody Molecules using 4-lodophenethylmaleimide Decreases Renal Uptake of Radioactivity"; Chemitry Open, vol. 4, pp. 174-182, 2015.
Strohl, William R., "Optimization of Fe-mediated effector functions of monoclonal antibodies," Current Opinion in Biotechnology, vol. 20, pp. 685-691 (2009).
SwissProt Accession No. P00533.2, "Epidermal Growth Factor Receptor," pp. 1-49 (Jun. 11, 2014).
Takahashi et al., "Cutting Edge: 4-1 BB Is a Bona Fide COB T Cell Survival Signal," J Immunol., vol. 162, pp. 0037-5040 (1999).
Tang et al, "Anti-Transferrin Receptor-Modified Amphotericin B-Loaded PLA-PEG Nanoparticles Cure Candidal Meningitis and Reduce Drug Toxicity," Oct. 5, 2015, International Journal of Medicine, 2015:10, pp. 6227-6241.
Tannock and Hill. The Basic Science of Oncology. 1998. New York: McGraw-Hill;; pp. 357-358.
Tie et al., "Safety and efficacy of nivolumab in the treatment of cancers: A meta-analysis of 27 prospective clinical rials," International Journal of Cancer, vol. 140, pp. 948-958, (2017).
Turke et al., "Preexistence and Clonal Selection of MET Amplification in EGFR Mutant NSCLC," Cancer Cell, vol. 17, pp. 77-88 (2010).
Ullrich et al., "Human epidermal growth factor receptor cDNA sequence and aberrant expression of the amplified Jene in A431 epidermoid carcinoma cells," Nature, vol. 309, pp. 418-425 (1984).
UniProt Accession No. P10039, Nov. 1, 1997.
Vajdos et al., "Comprehensive funtional maps of the antigen-binding site of an anti-ErbB2 antibody obtained with shotgun scanning mutagenisis", J. Mol. Biol. (2002) 32(2): pp. 415-428.
Van den Burg et al., "Selection of mutations for increased protein stability", Curr. Opin. Biotech. 13:333-337 (2002).
Wang et al., "VISTA, a novel mouse Ig superfamily ligand that negatively regulates T cell responses," Journal of Experimental Medicine, vol. 208, No. 3, pp. 577-592 (Mar. 14, 2011).
Watanabe et al., "Gene Cloning of Chitinase A1 from Bacillus circulans WL-12 Revealed Its Evolutionary Relationship to Serratia Chitinase and to the Type III Homology Units of Fibronectin," Journal of Biological Chemistry, vol. 265, pp. 15659-15665 (1990).
Wattanachaisaereekul, "Production of Polyketides by Saccharomyces cerevisiae", Ph.D. Thesis (2007) Center for Microbial Biotechnology, BioCentrum-DTU Technical University of Denmark, pp. 1-187.
Xu, et al., "Directed Evolution of High-Affinity Antibody Mimics Using mRNA Display,"Chemistry & Biology, 9: 933-942 (2002).
Zhang et al., "Complete disulfide bond assignment of a recombinant immunoglobulin G4 monoclonal antibody," Analytical Biochemistry, vol. 311, pp. 1-9 (2002).
Zhou et al., Characterization of human homologue of 4-1 BB and its ligand, Immunology Letters, vol. 45, pp. p7-73, 1995.
Zucali, et al.," Role of cMET expression in non-small-cell lung cancer patients treated with eGFR tyrosine kinase inhibitors", Annals of Anocology (2008) 19:: 1605-1612.

Also Published As

Publication number Publication date
US20180162928A1 (en) 2018-06-14
US10597438B2 (en) 2020-03-24
EP3554535A4 (en) 2020-10-21
US20210024612A1 (en) 2021-01-28
EP3554535A1 (en) 2019-10-23
WO2018111976A1 (en) 2018-06-21

Similar Documents

Publication Publication Date Title
US20210147510A1 (en) Prostate specific membrane antigen binding fibronectin type iii domains
US11447539B2 (en) PD-L1 binding fibronectin type III domains
US11345739B2 (en) CD137 binding fibronectin type III domains
US11628222B2 (en) CD71 binding fibronectin type III domains
US20190184028A1 (en) Targeting with firbronectin type iii like domain molecules
US20220332795A1 (en) Cd71 binding fibronectin type iii domains
US20230330239A1 (en) Epcam binding fibronectin type iii domains
CN117616045A (en) Fibronectin type III domain binding CD71
EA042392B1 (en) TYPE III FIBRONECTIN DOMAIN BINDING TO PROSTATE-SPECIFIC MEMBRANE ANTIGEN

Legal Events

Date Code Title Description
AS Assignment

Owner name: JANSSEN BIOTECH, INC., PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DIEM, MICHAEL;HAWKINS, REBECCA;JACOBS, STEVEN;AND OTHERS;SIGNING DATES FROM 20180906 TO 20181016;REEL/FRAME:051938/0865

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE